1
|
Lei Y, Zhou R, Mao Q, Qiu X, Mu D. The roles of pleiotrophin in brain injuries: a narrative review of the literature. Ann Med 2025; 57:2452353. [PMID: 39829367 PMCID: PMC11749013 DOI: 10.1080/07853890.2025.2452353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Pleiotrophin (PTN), a secreted multifunctional growth factor, is highly expressed in the developing brain. Recently, many studies have indicated that PTN participates in the development of brain and plays a neuroprotection after brain injury, especially promoting neuronal survival and neurite outgrowth, stimulating oligodendrocyte maturation and myelination, modulating neuroinflammation, and so on. OBJECTIVE However, no reviews comprehensively summarize the roles of PTN in brain injuries. Considering this, this review focuses on the roles and related regulatory pathways of PTN in brain injuries, what is known to date. METHODS PubMed and Embase databases have been searched, and related studies are compiled and summarized. RESULTS Our review has found PTN participates in the repairment of brain injuries, including hypoxic-ischemic brain injury, preterm white matter injury, traumatic brain injury, and neurodegenerative diseases, mainly based on animal data and small sample size studies. Besides, PTN interacts with receptors, such as, Z-type protein tyrosine phosphatase receptor and syndecan-3, regulating related pathways in these events. CONCLUSION It suggests PTN as a promising candidate for the treatment of brain injuries clinically. However, the evidence is early in its development. Further multi-center and large-sample studies are warranted to support our findings and determine the clinical value of PTN for treating brain injuries.
Collapse
Affiliation(s)
- Yupeng Lei
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Qian Mao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
2
|
Zhong X, Tai W, Liu ML, Ma S, Shen T, Zou Y, Zhang CL. The Citron homology domain of MAP4Ks improves outcomes of traumatic brain injury. Neural Regen Res 2025; 20:3233-3244. [PMID: 39314140 PMCID: PMC11881717 DOI: 10.4103/nrr.nrr-d-24-00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/19/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00027/figure1/v/2024-12-20T164640Z/r/image-tiff The mitogen-activated protein kinase kinase kinase kinases (MAP4Ks) signaling pathway plays a pivotal role in axonal regrowth and neuronal degeneration following insults. Whether targeting this pathway is beneficial to brain injury remains unclear. In this study, we showed that adeno-associated virus-delivery of the Citron homology domain of MAP4Ks effectively reduces traumatic brain injury-induced reactive gliosis, tauopathy, lesion size, and behavioral deficits. Pharmacological inhibition of MAP4Ks replicated the ameliorative effects observed with expression of the Citron homology domain. Mechanistically, the Citron homology domain acted as a dominant-negative mutant, impeding MAP4K-mediated phosphorylation of the dishevelled proteins and thereby controlling the Wnt/β-catenin pathway. These findings implicate a therapeutic potential of targeting MAP4Ks to alleviate the detrimental effects of traumatic brain injury.
Collapse
Affiliation(s)
- Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meng-Lu Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
You X, Niu L, Fu J, Ge S, Shi J, Zhang Y, Zhuang P. Bidirectional regulation of the brain-gut-microbiota axis following traumatic brain injury. Neural Regen Res 2025; 20:2153-2168. [PMID: 39359076 PMCID: PMC11759007 DOI: 10.4103/nrr.nrr-d-24-00088] [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: 01/23/2023] [Revised: 03/20/2024] [Accepted: 05/11/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00002/figure1/v/2024-09-30T120553Z/r/image-tiff Traumatic brain injury is a prevalent disorder of the central nervous system. In addition to primary brain parenchymal damage, the enduring biological consequences of traumatic brain injury pose long-term risks for patients with traumatic brain injury; however, the underlying pathogenesis remains unclear, and effective intervention methods are lacking. Intestinal dysfunction is a significant consequence of traumatic brain injury. Being the most densely innervated peripheral tissue in the body, the gut possesses multiple pathways for the establishment of a bidirectional "brain-gut axis" with the central nervous system. The gut harbors a vast microbial community, and alterations of the gut niche contribute to the progression of traumatic brain injury and its unfavorable prognosis through neuronal, hormonal, and immune pathways. A comprehensive understanding of microbiota-mediated peripheral neuroimmunomodulation mechanisms is needed to enhance treatment strategies for traumatic brain injury and its associated complications. We comprehensively reviewed alterations in the gut microecological environment following traumatic brain injury, with a specific focus on the complex biological processes of peripheral nerves, immunity, and microbes triggered by traumatic brain injury, encompassing autonomic dysfunction, neuroendocrine disturbances, peripheral immunosuppression, increased intestinal barrier permeability, compromised responses of sensory nerves to microorganisms, and potential effector nuclei in the central nervous system influenced by gut microbiota. Additionally, we reviewed the mechanisms underlying secondary biological injury and the dynamic pathological responses that occur following injury to enhance our current understanding of how peripheral pathways impact the outcome of patients with traumatic brain injury. This review aimed to propose a conceptual model for future risk assessment of central nervous system-related diseases while elucidating novel insights into the bidirectional effects of the "brain-gut-microbiota axis."
Collapse
Affiliation(s)
- Xinyu You
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Niu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiafeng Fu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shining Ge
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangwei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanjun Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Pengwei Zhuang
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
4
|
Filho WGDS, do Nascimento Moura LJ, Nascimento ABM, Tessmann GC, Miranda FS, de Almeida VCR, Broedel BV, de Faria ML, Arêas FZDS. Electroencephalogram biomarkers as predictors of mortality and functional recovery in patients with severe traumatic brain injury: Protocol study. MethodsX 2025; 14:103146. [PMID: 39877472 PMCID: PMC11773473 DOI: 10.1016/j.mex.2024.103146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Traumatic brain injury (TBI) is a global public health condition that causes cognitive and behavioral deficits. This protocol assesses the potential of quantitative electroencephalogram (EEG) biomarkers, associated with inflammatory indicators, to predict mortality and functional recovery in patients with severe TBI. Through continuous monitoring and analysis of abnormal brain activity patterns, the protocol aims to personalize therapeutic interventions and improve patient quality of life. This randomized clinical trial includes 84 adult participants with severe TBI, followed at different stages of recovery, using validated scales for functional and predictive analysis. Traumatic brain injury (TBI) is a globally impactful public health condition characterized by initial brain injuries caused by traumatic forces, leading to cognitive and behavioral deficits. The trauma triggers inflammatory and neurochemical changes that exacerbate neuronal damage, resulting in neuropsychiatric complications. The use of electroencephalogram (EEG), particularly in its quantitative form (QEEG), is crucial for patients with severe TBI, as it allows early detection of abnormal brain activity patterns, such as slow waves, which indicate a worse prognosis. This continuous monitoring, combined with inflammatory biomarkers, guides personalized therapeutic interventions and improves the prediction of clinical outcomes, contributing to patient quality of life.
Collapse
Affiliation(s)
- Walter Gomes da Silva Filho
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
- Postgraduate program in Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Layza Julhia do Nascimento Moura
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Arthur Barcelos Massariol Nascimento
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Gabrielle Cristina Tessmann
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Fabricia Silva Miranda
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Vitória Caroline Reinoso de Almeida
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Bárbara Vargens Broedel
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Miller Lucas de Faria
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
| | - Fernando Zanela da Silva Arêas
- Neurorehabilitation and Neuromodulation Laboratory, Department of Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
- Postgraduate program in Physiological Sciences, Federal University of Espírito Santo, City of Vitória, ES, Brazil
- Baylor Scott and White Research Institute and Baylor Scott and White Institute for Rehabilitation, Dallas, TX, USA
| |
Collapse
|
5
|
Sun Y, Gao SQ, Wang X, Li T, Han YL, Miao SH, Zhao R, Zheng XB, Qiu JY, Jin WX, Gao CC, Zhou ML. Galectin-3 activates microglia and promotes neurological impairment via NLRP3/pyroptosis pathway following traumatic brain injury. Brain Res 2025; 1855:149560. [PMID: 40074166 DOI: 10.1016/j.brainres.2025.149560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Externally caused traumatic brain injury (TBI) poses a woeful worldwide health concern, bringing about disability, death, and prolonged neurological impairment. Increased galectin-3 levels have been linked to unfavorable outcomes in several neurological conditions. This study explores the role of galectin-3 in TBI, specifically examining its contribution to neuroinflammation. METHODS BV2 microglia cells treated with lipopolysaccharide (LPS) and a mouse model of TBI were applied to investigate the impact of galectin-3 on neuroinflammation following TBI. Western blotting and immunofluorescence labeling were applied for evaluating protein levels and colocalization. Adeno-associated virus (AAV) that targets microglia was used to knock down galectin-3 in microglia. Nissl staining and the modified neurologic severity score were employed in evaluating neural survival and neurological function, and the cognitive impairment following TBI was assessed by the Y-Maze and Morri water maze test. RESULTS Galectin-3 expression was shown to rise dramatically after TBI, peaking between days five and seven. In vitro, BV2 cells treated with LPS showed reduced NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome activation when galectin-3 was inhibited. In LPS-activated microglia, galectin-3 inhibition specifically decreased the expression of Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), p-NF-κB, NLRP3, Apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, and Gasdermin D (GSDMD). Injection with AAV containing siRNA to knock down galectin-3 in microglia was operated on mice in vivo. Following TBI, this knockdown led to reduced NLRP3 inflammasome activation, neuronal death, neurological impairments and cognitive impairment. CONCLUSIONS Our foundings indicate that modulating microglia-derived galectin-3 following TBI to reduce neuroinflammation could serve as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Sheng-Qing Gao
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xue Wang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Li
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yan-Ling Han
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shu-Hao Miao
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ran Zhao
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiao-Bo Zheng
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing. China
| | - Jia-Yin Qiu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wang-Xuan Jin
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao-Chao Gao
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Meng-Liang Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
6
|
Magyar-Sumegi ZD, Csendes M, Lendvai-Emmert D, Sebestyen G, Tamas V, Bandi S, Czigler A, Yabluchanskiy A, Tarantini S, Ungvari Z, Czeiter E, Amrein K, Orsi G, Perlaki G, Buki A, Toth P. Chronic impairment of neurovascular coupling and cognitive decline in young survivors of severe traumatic brain injury. GeroScience 2025:10.1007/s11357-025-01683-w. [PMID: 40360822 DOI: 10.1007/s11357-025-01683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Severe traumatic brain injury (TBI) leads to chronic cognitive decline, imposing a significant societal burden. The regulation of cerebral blood flow (CBF) is critical for cognitive function, and acute disruptions in CBF regulation predict poor TBI outcomes. However, the long-term effects of TBI on CBF regulation and their association with cognitive function remain poorly understood. This study aimed to investigate whether severe TBI results in chronic CBF dysregulation and whether this contributes to long-term cognitive deficits. Additionally, we examined the role of TBI-induced insulin-like growth factor 1 (IGF-1) deficiency in cerebrovascular dysfunction. We assessed cognitive function, basal CBF (via phase contrast MRI), CBF autoregulation (via transcranial Doppler), and neurovascular coupling (NVC) in 33 TBI survivors (mean age 37.6 years, ~ 10 years post-injury) and 21 age-matched healthy controls. Serum IGF-1 levels were also measured. TBI survivors exhibited significant impairments in memory and executive function compared to controls. While basal CBF and autoregulation remained intact, NVC responses were chronically impaired and correlated with cognitive deficits. However, IGF-1 levels did not differ between groups and were not associated with NVC impairment or cognitive function. Our findings indicate that severe TBI results in chronic impairment of neurovascular coupling, which likely contributes to long-term cognitive deficits. These results highlight the need for further research to identify underlying neurovascular mechanisms and develop interventions to restore NVC and cognitive function in TBI survivors.
Collapse
Affiliation(s)
- Zsofia Dina Magyar-Sumegi
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Department of Psychiatry and Psychotherapy, Medical School, University of Pecs, Pecs, Hungary
- Doctoral School of Clinical Neurosciences, Medical School, University of Pecs, Pecs, Hungary
| | - Mark Csendes
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | | | - Gabriella Sebestyen
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Doctoral School of Clinical Neurosciences, Medical School, University of Pecs, Pecs, Hungary
| | - Viktoria Tamas
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Szabolcs Bandi
- Department of Psychiatry and Psychotherapy, Medical School, University of Pecs, Pecs, Hungary
| | - Andras Czigler
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College Health Sciences Division/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College Health Sciences Division/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College Health Sciences Division/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- HUN-REN-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
- Molecular Medicine Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Krisztina Amrein
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- HUN-REN-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
- Molecular Medicine Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Gergely Orsi
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Gabor Perlaki
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Andras Buki
- Department of Neurosurgery, Faculty of Medicine and Health, Orebro University, Orebro, Sweden
| | - Peter Toth
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary.
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Doctoral College Health Sciences Division/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
7
|
Gilfarb RA, Ranade S, Smail M, Wangler L, Stewart M, Rajesh A, Lenz KM, Leuner B. Hormonal contraceptives during adolescence impact the female brain and behavior in a rat model. Horm Behav 2025; 171:105725. [PMID: 40188588 DOI: 10.1016/j.yhbeh.2025.105725] [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: 10/15/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/08/2025]
Abstract
Millions of people take hormonal contraceptives (HCs), often starting during adolescence when ovarian hormones influence brain and behavioral maturation. However, there is a fundamental lack of information about the neurobehavioral consequences of hormonal alterations via adolescent HC use. To begin addressing this gap, we validated a rodent model of adolescent HC administration and characterized its impact on endocrine, transcriptional, and behavioral endpoints. Cohorts of intact post-pubertal female Sprague-Dawley rats received daily subcutaneous injections of either vehicle or HC [10 μg ethinyl estradiol (EE) + 20 μg levonorgestrel (LNG)] for the duration of adolescence from postnatal day (PND) 35 to PND56. Blood and brain tissue was collected at PND57. Other cohorts received daily injections of vehicle or HC from PND35 until behavioral assays were completed on PND57-64. HC treatment was effective, as vaginal lavage indicated disrupted estrous cycling and ELISA indicated suppressed serum luteinizing hormone in HC-treated rats. Liquid chromatography-mass spectrometry analysis showed EE and LNG in serum and brain as well as diminished serum and brain levels of allopregnanolone and testosterone in HC-treated rats. NanoString nCounter analysis indicated that adolescent HC administration impacted expression of genes related to synapses, white matter, neuroimmune, monoamine, and hormone signaling in the hypothalamus and medial prefrontal cortex. While no effects of HCs were seen on sociability in the social preference test or stress coping behavior in the forced swim test, adolescent HC administration diminished risk-assessment behaviors in the novelty-induced hypophagia paradigm and altered anxiety-like behavior in the open field test and elevated plus maze. Overall, these data suggest that exposure to contraceptive hormones during the critical developmental period of adolescence may shape the brain and behavior.
Collapse
Affiliation(s)
- Rachel A Gilfarb
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Sanjana Ranade
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Marissa Smail
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Lynde Wangler
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Meredith Stewart
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Abhishek Rajesh
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 370 W. 9th Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 370 W. 9th Avenue, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
8
|
Packer JM, Giammo SG, Wangler LM, Davis AC, Bray CE, Godbout JP. Diffuse traumatic brain injury induced stimulator of interferons (STING) signaling in microglia drives cortical neuroinflammation, neuronal dysfunction, and impaired cognition. J Neuroinflammation 2025; 22:128. [PMID: 40307881 PMCID: PMC12044788 DOI: 10.1186/s12974-025-03451-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Neuropsychiatric complications including depression and cognitive impairment develop, persist, and worsen in the years after traumatic brain injury (TBI), negatively affecting life and lifespan. Inflammatory responses mediated by microglia are associated with the transition from acute to chronic neuroinflammation after TBI. Moreover, type I interferon (IFN-I) signaling is a key mediator of inflammation during this transition. Thus, the purpose of this study was to determine the degree to which a microglia-specific knockout of the stimulator of interferons (STING) influenced TBI-induced neuroinflammation, neuronal dysfunction, and cognitive impairment. Here, microglial inducible STING knockout (CX₃CR1Cre/ERT2 x STINGfl/fl) mice were created and validated (mSTING-/-). Diffuse brain injury (midline fluid percussion) in male and female mice increased STING expression in microglia, promoted microglial morphological restructuring, and induced robust cortical inflammation and pathology 7 days post injury (dpi). These TBI-associated responses were attenuated in mSTING-/- mice. Increased cortical astrogliosis and rod-shaped microglia induced by TBI were independent of mSTING-/-. 7 dpi, TBI induced 237 differentially expressed genes (DEG) in the cortex of functionally wildtype (STINGfl/fl) associated with STING, NF-κB, and Interferon Alpha signaling and 85% were attenuated by mSTING-/-. Components of neuronal injury including reduced NeuN expression, increased cortical lipofuscin, and increased neurofilament light chain in plasma were increased by TBI and dependent on mSTING. TBI-associated cognitive tasks (novel object recognition/location, NOR/NOL) at 7 dpi were dependent on mSTING. Notably, the TBI-induced cognitive deficits in NOR/NOL and increased cortical inflammation 7 dpi were unaffected in global interferon-α/β receptor 1 knockout (IFNAR1) mice. In the final study, the RNA profile of neurons after TBI in STINGfl/fl and mSTING-/- mice was assessed 7 dpi by single nucleus RNA-sequencing. There was a TBI-dependent suppression of cortical neuronal homeostasis with reductions in CREB signaling, synaptogenesis, and oxytocin signaling and increases in cilium assembly and PTEN signaling. Overall, mSTING-/- prevented 50% of TBI-induced DEGs in cortical neurons. Collectively, ablation of STING in microglia attenuates TBI-induced interferon responses, cortical inflammation, neuronal dysfunction, neuronal pathology, and cognitive impairment.
Collapse
Affiliation(s)
- Jonathan M Packer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10Th Ave, Columbus, OH, USA
| | - Samantha G Giammo
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10Th Ave, Columbus, OH, USA
| | - Lynde M Wangler
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10Th Ave, Columbus, OH, USA
| | - Amara C Davis
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10Th Ave, Columbus, OH, USA
| | - Chelsea E Bray
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10Th Ave, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10Th Ave, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, 175 Pomerene Hall, Columbus, OH, USA.
- 231 IBMR Building, The Ohio State University, 460 Medical Center Dr., Columbus, OH, 43210, USA.
| |
Collapse
|
9
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025; 19:15189-15219. [PMID: 40249331 PMCID: PMC12045021 DOI: 10.1021/acsnano.5c00700] [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: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
10
|
Liu X, Xia J, Shao W, Li X, Yuan D, Xie J, Zhang L, Tang Y, Zhao H, Wu P. Adhesion-Related Pathways and Functional Polarization of Astrocytes in Traumatic Brain Injury: Insights from Single-cell RNA Sequencing. Neuromolecular Med 2025; 27:30. [PMID: 40287916 DOI: 10.1007/s12017-025-08858-w] [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: 03/06/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Traumatic brain injury (TBI) induces profound functional heterogeneity in astrocytes, yet the regulatory mechanisms underlying this diversity remain poorly understood. In this study, we analyzed single-cell RNA sequencing data from the cortex and hippocampus of TBI mouse models to characterize astrocyte subtypes and their functional dynamics. We identified two major reactive subtypes: A1 astrocytes, enriched in inflammatory response, synaptic regulation, and neurodegenerative disease-related pathways; and A2 astrocytes, enriched in lipid metabolism, extracellular matrix (ECM) remodeling, and phagosome formation pathways. These functional differences were consistently observed across datasets with varying injury severities. Notably, adhesion-related pathways-including gap junctions, adherens junctions, and calcium-dependent adhesion-showed significant subtype-specific expression patterns and temporal shifts. Pseudotime trajectory analysis further suggested a potential transition between A1 and A2 states, accompanied by dynamic regulation of adhesion-related genes. Our findings highlight the complex and context-dependent roles of astrocytes in TBI and propose cell adhesion as a key modulator of astrocyte functional polarization.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Ji Xia
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Wenjing Shao
- Department of Anesthesiology, Chongqing Huamei Plastic Surgery Hospital, Chongqing, 400015, China
| | - Xiaoming Li
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Danfeng Yuan
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Jingru Xie
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Liang Zhang
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Yuqian Tang
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Hui Zhao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Pengfei Wu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China.
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China.
| |
Collapse
|
11
|
Wang L, Guo H, Zhao W, Wang J, Cao X. Oxiracetam ameliorates neurological function after traumatic brain injury through competing endogenous RNA regulatory network. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06797-9. [PMID: 40272502 DOI: 10.1007/s00213-025-06797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
RATIONALE Oxiracetam (ORC) has been demonstrated to improve neurological function resulting from traumatic brain injury (TBI). OBJECTIVES This study aims to explore the precise molecular mechanism of ORC in the treatment of TBI. METHODS TBI rat model was established and treated with ORC. Modified Garcia score, rotarod test and HE staining were employed to evaluate the neuroprotective effects of ORC. Subsequently, RNA-seq was conducted on the hippocampus of sham, TBI and ORC rats to identify differential expression (DE) lncRNAs and mRNAs. Functional analysis of DE lncRNAs and mRNAs was performed. The real-time quantitative polymerase chain reaction (qRT-PCR) was used to determine the expression of DE lncRNAs and DE mRNAs. Western blot was performed to explore important pathway in ceRNA networks. RESULTS ORC has been demonstrated to effectively improve neurological function in TBI rats. A total of 10 ORC-treated DE lncRNAs and 61 DE mRNAs were obtained. A co-expression network comprising 79 lncRNA-mRNA pairs associated with the treatment of ORC was constructed. Furthermore, an lncRNA-miRNA-mRNA regulated ceRNA network was constructed, comprising 15 mRNAs, 41 miRNAs and 10 lncRNAs. Functional enrichment, qRT-PCR, and Western blot analysis showed that ORC improve neurological function of TBI rats by regulating multiple signaling pathways, including the JAK-STAT/PI3K-Akt pathway, as well as affecting the expression of key genes Prlr, Cdkn1a, and Cldn1. CONCLUSION Our study reveals the mechanism of ORC therapy in TBI rats, which mainly relies on the regulation of the JAK-STAT/PI3K-Akt pathway and the influence on the expression of key genes Prlr, Cdkn1a, and Cldn1.
Collapse
Affiliation(s)
- Liyi Wang
- Hospital Infection-Control Department, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Han Guo
- Department of Oral Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Weidong Zhao
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Jiahao Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Xuhua Cao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, 050000, China.
| |
Collapse
|
12
|
Wang M, Zhao R, Su Y, Zhai D, Liang H, Zhang L, Wang W, Wang Z, Qi M, Jiang X, Ling S, Di G. 4,4'-Dimethoxychalcone Mitigates Neuroinflammation Following Traumatic Brain Injury Through Modulation of the TREM2/PI3K/AKT/NF-κB Signaling Pathway. Inflammation 2025:10.1007/s10753-025-02279-4. [PMID: 40261458 DOI: 10.1007/s10753-025-02279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/24/2025]
Abstract
Research on 4,4'-dimethoxychalcone (DMC) in the context of traumatic brain injury (TBI) is extremely limited, and no effective clinical treatments are available to improve outcomes for individuals with TBI. Our study aims to investigate the underlying mechanisms by which DMC may alleviate neuroinflammation and neuronal damage following TBI. This study seeks to provide a theoretical foundation for the development of future pharmacological therapies for TBI. A moderate TBI model was established using the fluid percussion injury (FPI) method. The recovery of neuromotor function following TBI was evaluated using the modified neurological severity score (mNSS), the Morris water maze test, and analysis of cerebral edema. Gene and protein expression levels were quantified using cell viability assays, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and immunofluorescence. Network pharmacology was employed to predict potential targets of DMC, and gene ontology (GO) analysis along with KEGG pathway enrichment was conducted to predict signaling pathways affected by DMC.DMC treatment significantly improved neuromotor deficits in mice after TBI. In both in vivo and in vitro experiments, DMC suppressed microglial activation and decreased the production and release of inflammatory factors. Additionally, DMC reduced neuronal lesions after TBI. DMC notably decreased the elevated expression of triggering receptor expressed on myeloid cells 2 (TREM2) following TBI. Network pharmacological analysis indicated that DMC's therapeutic effects may be mediated through the PI3K/AKT signaling cascade. These findings indicate that DMC has therapeutic potential for TBI, with significant anti-inflammatory and neuroprotective properties likely mediated by the TREM2/PI3K/AKT/NF-κB signaling cascade.
Collapse
Affiliation(s)
- Mengran Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Rui Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Yue Su
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Duhuan Zhai
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Hengyan Liang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Lingkun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Weicheng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Zhichun Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Min Qi
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Xiaochun Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China.
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
| | - Shizhang Ling
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China.
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
| | - Guangfu Di
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China.
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
| |
Collapse
|
13
|
Medeiros-Furquim T, Miedema A, Schilder E, Brouwer N, Holtman IR, Kooistra SM, Eggen BJL. Microglia endotoxin tolerance is retained after enforced repopulation. Brain Behav Immun 2025; 128:512-528. [PMID: 40274001 DOI: 10.1016/j.bbi.2025.04.014] [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: 07/24/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Microglia are crucial for CNS homeostasis and are involved in a wide range of neurodegenerative and neuroinflammatory diseases. Systemic inflammation and infections can contribute to neurodegeneration later in life by affecting microglia. Like other innate immune cells, microglia can develop innate immune memory (IIM) in response to an inflammatory challenge, altering their response to subsequent stimuli. IIM can ameliorate or worsen CNS pathology, but it is unclear if IIM can be reversed to restore microglia functions. Here, we investigated whether microglia depletion-repopulation by inhibition of the colony-stimulating factor 1 receptor with BLZ945 reversed LPS-induced microglia endotoxin tolerance in mice. Repopulated microglia displayed a reduced expression of homeostatic genes and genes related to mitochondrial respiration and TCA cycle metabolism and an increased expression of immune effector and activation genes. Nonetheless, the blunted inflammatory gene response after LPS-preconditioning was retained after a depletion-repopulation cycle. Our study highlights the persistence of endotoxin tolerance in microglia after a depletion-repopulation cycle, which might impact the potential effectiveness of strategies targeted at microglia depletion for clinical applications.
Collapse
Affiliation(s)
- Tiago Medeiros-Furquim
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anneke Miedema
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Edwin Schilder
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge R Holtman
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Susanne M Kooistra
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Bart J L Eggen
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
14
|
Zuo L, Geng L, Cao Y, Zhou XY, Di W, Liu Y, Zhong Z, Liu D, Zhang Z, Yan F. Circulating Neutrophil-to-Lymphocyte Ratio Predicts Stroke-Associated Infection and Poststroke Fatigue Affecting Long-Term Neurological Outcomes in Stroke Patients. Mediators Inflamm 2025; 2025:5202480. [PMID: 40308934 PMCID: PMC12041617 DOI: 10.1155/mi/5202480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/22/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Since peripheral leukocytes may contribute to the pathophysiology of stroke, the aim of this study was to elucidate the relationship between leukocytes and stroke outcomes and identify which leukocyte subtypes most accurately predict functional outcomes and poststroke fatigue (PSF) in stroke patients. Methods: A total of 788 ischemic stroke patients within 72 h of onset of disease were admitted in our study. Stroke-associated infection (SAI) and PSF were evaluated according to diagnosis standards by a special neurologist. Analyses were performed using SPSS 23.0 and GraphPad Prism 10.0. Results: Neutrophil-to-lymphocyte ratio (NLR) has discriminative power in predicting stroke outcome, and the area under the curve (AUC) of NLR to distinguish stroke outcomes was 0.689 (95% confidence interval, 0.646-0.732). Positive correlation was found between NLR levels and NIHSS score on admission (r = 0.2786, p < 0.001). Risk model for predicting stroke outcome was constructed using age, NIHSS, previous stroke history, triglycerides, glucose and hemoglobin levels, thrombolysis treatment, and NLR, with an AUC of 0.865. Patients who developed SAI and PSF both had significantly higher NLR levels at admission than those patients not diagnosed with SAI and PSF (p < 0.0001). A risk model was constructed to predict PSF based on parameters including age, NIHSS score, lipoprotein(a) and NLR, and an AUC of 0.751. Conclusions: Higher NLR levels in the acute phase of stroke might indicate a higher incidence of SAI and PSF. Therefore, higher NLR is associated with a poor stroke prognosis.
Collapse
Affiliation(s)
- Lei Zuo
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Leiyu Geng
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Yujia Cao
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Xin-yu Zhou
- Department of Neurology, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Wu Di
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Yun Liu
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Zhe Zhong
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Dandan Liu
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Zhengsheng Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| | - Fuling Yan
- Department of Neurology, Affiliated ZhongDa Hospital, Medical school of Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
15
|
Zhang Y, He K, Zhang C, Dang H, Hei J, Zhang Y, Chen P, Zhang Z, Yang Y, Wang Z, Yang X, Zhang L, Yu Y. Atlas of temporal molecular pathological alterations after traumatic brain injury based on RNA-Seq. Exp Neurol 2025; 390:115270. [PMID: 40268159 DOI: 10.1016/j.expneurol.2025.115270] [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: 12/22/2024] [Revised: 04/01/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
Traumatic brain injury (TBI) involves diverse molecular pathological alterations and biological processes in a temporally dynamic manner. However, current knowledge on the various processes during the acute phase of TBI is still rather limited. RNA-seq analysis was performed on brain tissues from C57/BL6 mice at 10 time points(0 h, 1 h, 2 h, 3 h, 4 h, 6 h, 12 h, 1d, 3d, and 7d) following TBI modeling. Subsequently, a bioinformatics approach, Weighted Gene Co-expression Network Analysis (WGCNA), was employed to identify characteristic modules, which were then validated using the Mfuzz method. Pathway enrichment analysis was conducted on WGCNA module genes, and hub genes were screened using the STRING database. After exploring the various potential pathways and expression patterns (neuroinflammation, cognition, gliosis and myelin regeneration etc.), we focus on pyroptosis, a inflammatory cell death influencing immune response, for in-depth analysis. RT-qPCR, Western blot(WB) and Immunofluorescence(IF) were used to validate the hub genes and key pyroptosis-related genes(Casp1, Casp11, GSDMD). Additionally, single-cell RNA sequencing data at 7 day post injury(dpi) was also used to validate the expression of the identified hub genes. Our approach to intensive transcriptomic analysis comprehensively reveals the temporal molecular pathological alterations during TBI progression. Pyroptosis may be a key mechanism in the neuroinflammatory process. Intervention strategies targeting specific molecular pathways may offer novel approach for the treatment of TBI.
Collapse
Affiliation(s)
- Yulian Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Kun He
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Chuanpeng Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Hanhan Dang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Junru Hei
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Yunsheng Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Pengyu Chen
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Ze Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Yanbo Yang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Zixi Wang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Xu Yang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Li Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Yanbing Yu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China.
| |
Collapse
|
16
|
Holcomb M, Marshall AG, Flinn H, Lozano-Cavazos M, Soriano S, Gomez-Pinilla F, Treangen TJ, Villapol S. Probiotic treatment induces sex-dependent neuroprotection and gut microbiome shifts after traumatic brain injury. J Neuroinflammation 2025; 22:114. [PMID: 40254574 PMCID: PMC12010691 DOI: 10.1186/s12974-025-03419-1] [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: 03/31/2024] [Accepted: 03/16/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Recent studies have highlighted the potential influence of gut dysbiosis on traumatic brain injury (TBI) outcomes. Alterations in the abundance and diversity of Lactobacillus species may affect immune dysregulation, neuroinflammatory responses, anxiety- and depressive-like behaviors, and neuroprotective mechanisms activated in response to TBI. OBJECTIVE This study aims to evaluate the protective and preventive effects of Pan-probiotic (PP) treatment on the inflammatory response during both the acute and chronic phases of TBI. METHODS Males and female mice underwent controlled cortical impact (CCI) injury or sham. They received a PP mixture in drinking water containing strains of Lactobacillus plantarum, L. reuteri, L. helveticas, L. fermentum, L. rhamnosus, L. gasseri, and L. casei. In the acute group, mice received PP or vehicle (VH) treatment for 7 weeks before TBI, continuing until 3 days post-injury (dpi). In the chronic group, treatment began 2 weeks before TBI and was extended through 35 dpi. The taxonomic microbiome profiles of fecal samples were evaluated using 16S rRNA V1-V3 sequencing analysis, and Short-chain fatty acids (SCFAs) were measured. Immunohistochemical, in situ hybridization, and histological analyses were performed to assess neuroinflammation post-TBI, while behavioral assessments were conducted to evaluate sensorimotor and cognitive functions. RESULTS Our findings suggest that a 7-week PP administration induces specific microbial changes, including increased abundance of beneficial bacteria such as Lactobacillaceae, Limosilactobacillus, and Lactiplantibacillus. PP treatment reduces lesion volume and cell death at 3 dpi, elevates SCFA levels at 35 dpi, and decreases microglial activation at both time points, particularly in males. Additionally, PP treatment improved motor recovery in males and alleviated depressive-like behaviors in females. CONCLUSION Our findings indicate that PP administration modulates microbiome composition, reduces neuroinflammation, and improves motor deficits following TBI, with these effects being particularly pronounced in male mice.
Collapse
Affiliation(s)
- Morgan Holcomb
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Austin G Marshall
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Hannah Flinn
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Mariana Lozano-Cavazos
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Sirena Soriano
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Fernando Gomez-Pinilla
- Departments of Neurosurgery and Integrative Biology and Physiology, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Ken Kennedy Institute, Rice University, Houston, TX, USA
| | - Sonia Villapol
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York City, NY, USA.
| |
Collapse
|
17
|
Kerr NA, Choi J, Mohite SY, Singh PK, Bramlett HM, Lee JK, Dietrich WD. Single cell RNA sequencing after moderate traumatic brain injury: effects of therapeutic hypothermia. J Neuroinflammation 2025; 22:110. [PMID: 40251570 PMCID: PMC12007139 DOI: 10.1186/s12974-025-03430-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/25/2025] [Indexed: 04/20/2025] Open
Abstract
Traumatic brain injury (TBI) initiates a cascade of cellular and molecular events that promote acute and long-term patterns of neuronal, glial, vascular, and synaptic vulnerability leading to lasting neurological deficits. These complex responses lead to patterns of programmed cell death, diffuse axonal injury, increased blood-brain barrier disruption, neuroinflammation, and reactive gliosis, each a potential target for therapeutic interventions. Posttraumatic therapeutic hypothermia (TH) has been reported to be highly protective after brain and spinal cord injury and studies have investigated molecular mechanisms underlying mild hypothermic protection while commonly assessing heterogenous cell populations. In this study we conducted single-cell RNA sequencing (scRNA-seq) on cerebral cortical tissues after experimental TBI followed by a period of normothermia or hypothermia to comprehensively assess multiple cell type-specific transcriptional responses. C57BL/6 mice underwent moderate controlled cortical impact (CCI) injury or sham surgery and then placed under sustained normothermia (37⁰C) or hypothermia (33⁰C) for 2 h. After 24 h, cortical tissues including peri-contused regions were processed for scRNA-seq. Unbiased clustering revealed cellular heterogeneity among glial and immune cells at this subacute posttraumatic time point. The analysis also revealed vascular and immune subtypes associated with neovascularization and debris clearance, respectively. Compared to normothermic conditions, TH treatment altered the abundance of specific cell subtypes and induced reactive astrocyte-specific modulation of neurotropic factor gene expression. In addition, an increase in the proportion of endothelial tip cells in the hypothermic TBI group was documented compared to normothermia. These data emphasize the importance of early temperature-sensitive glial and vascular cell processes in producing potentially neuroprotective downstream signaling cascades in a cell-type-dependent manner. The use of scRNA-seq to address cell type-specific mechanisms underlying therapeutic treatments provides a valuable resource for identifying targetable biological pathways for the development of neuroprotective and reparative interventions.
Collapse
Affiliation(s)
- Nadine A Kerr
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - James Choi
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simone Y Mohite
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Praveen Kumar Singh
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Helen M Bramlett
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Bruce W. Carter Department of Veterans Affairs Center, Miami, FL, USA
| | - Jae K Lee
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Neurological Surgery, University of Miami School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA.
| |
Collapse
|
18
|
Zhang B, Bai M, Yang M, Wang Y, Zhang X, Chen X, Gao M, Liu B, Shi G. Balancing Anti-Inflammation and Neurorepair: The Role of Mineralocorticoid Receptor in Regulating Microglial Phenotype Switching After Traumatic Brain Injury. CNS Neurosci Ther 2025; 31:e70404. [PMID: 40277259 PMCID: PMC12023002 DOI: 10.1111/cns.70404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND As potent anti-inflammatory agents, glucocorticoids (GCs) have been widely used in the treatment of traumatic brain injury (TBI). However, their use remains controversial. Our previous study indicated that although dexamethasone (DEX) exerted anti-inflammatory effects and protected the blood-brain barrier (BBB) by activating the glucocorticoid receptor (GR) after TBI, it also impeded tissue repair processes due to excessive anti-inflammation. Conversely, fludrocortisone, acting as a specific mineralocorticoid receptor (MR) agonist, has shown potential in controlling neuroinflammation and promoting neurorepair, but the underlying mechanisms need further exploration. OBJECTIVE This study aimed to explore the impact of the MR agonist fludrocortisone on microglia polarization, angiogenesis, functional rehabilitation, and associated mechanisms after TBI. METHODS We established a mice controlled cortical impact model, and then immunofluorescence staining, western blot, rt-PCR, and MRI were performed to investigate microglia polarization, angiogenesis, and brain edema in the ipsilateral hemisphere after TBI and fludrocortisone treatment. Subsequently, functional tests including morris water maze, sucrose preference test, and forced swimming test were conducted to evaluate the effects of fludrocortisone treatment on neurofunction after TBI. RESULTS Our results revealed that fludrocortisone suppressed neuroinflammation, enhanced angiogenesis and neuronal survival, and promoted functional rehabilitation by inducing a shift in microglia phenotype from M1 to M2 via the JAK/STAT6/PPARγ pathway. Additionally, the PI3K/Akt/HIF-1α pathway was involved in VEGF expression and in the process of angiogenesis. CONCLUSION Fludrocortisone, the specific MR agonist, exerted anti-neuroinflammatory and neuroprotective effects by regulating phenotypic switching of microglia from M1 to M2 rather than suppressing all types of microglia. Our study provided a theoretical basis for the therapeutic strategy of GCs targeting neuroinflammation after TBI.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Critical Care Medicine, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Miao Bai
- Department of NeurologyThe First Hospital of Tsinghua UniversityBeijingChina
| | - Mengshi Yang
- Department of Critical Care Medicine, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yumei Wang
- Department of Critical Care Medicine, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xueling Zhang
- Department of Critical Care Medicine, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xiyu Chen
- Department of Critical Care Medicine, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Min Gao
- Department of Animal LaboratoryBeijing Neurosurgical InstituteBeijingChina
| | - Baiyun Liu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Guangzhi Shi
- Department of Critical Care Medicine, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
19
|
Cheng Y, Gu W, Wu X, Tian W, Mu Z, Ye Y, Chao H, Bao Z. Allicin alleviates traumatic brain injury-induced neuroinflammation by enhancing PKC-δ-mediated mitophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156500. [PMID: 39986225 DOI: 10.1016/j.phymed.2025.156500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/21/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Traumatic brain injury (TBI) leads to neuroinflammation, which is a key contributor to the negative prognosis in TBI patients. Recent evidence indicates that allicin can prevent neuronal injury after TBI. However, whether allicin alleviates neuroinflammation by promoting mitophagy is unclear. PURPOSE We investigated the suppressive effects of allicin on neuroinflammation and clarified the role of mitophagy in the underlying mechanism. STUDY DESIGN/METHODS The controlled cortical impact (CCI) was employed to effectively mimic TBI in a living system. Cellular mechanical damage was modeled in vitro using a Bv2 cell stretch model. Neuroinflammation was assessed by evaluating levels of TNF-α, IL-1β, IL-6, ROS, IL-4 and IL-10, along with the expression of NLRP3 and TLR4 proteins. RNA-sequence and KEGG analyses revealed allicin-regulated molecular processes in the Bv2 cell stretch model. Immunofluorescence staining was performed to label both the autophagy marker protein LC3 and the outer mitochondrial membrane (OMM) marker COX IV. Lipid MS and lipidomic analyses were used to determine the CL levels in the OMM and IMM. The characteristic bilayer structure of mitochondria was observed using transmission electron microscopy (TEM). PKC-δ expression and phosphorylated phospholipid scramblase-3 (PLS3) levels were detected via western blotting. Stretched Bv2 cells and primary neurons were cocultured to assess the anti-neuroinflammatory effects of allicin. Neuro-rehabilitation was assessed using behavioral experiments such as the rotarod and morris water maze (MWM) tests. RESULTS Allicin treatment reduced TNF-α, IL-1β, IL-6, ROS levels, and the expression of NLRP3 and TLR4 proteins in mice with CCI, while IL-4 and IL-10 levels remained unchanged. Additionally, allicin reduced tissue lesions and cell death after CCI. The transcriptomic analysis revealed that mitophagy was important in allicin-related molecular pathways. The translocation of CL from IMM to OMM was facilitated by allicin, as demonstrated by flow cytometry and lipidomic analyses. Importantly, allicin increased PKC-δ expression and PLS3 phosphorylation in the CL-related mitophagy process in both the CCI and Bv2 cell stretch models. These findings suggest that allicin reduces mitophagy-related neuroinflammation and further prevents neuronal injury in vitro. Rottlerin, a selective PKC-δ inhibitor, effectively diminished allicin's capacity to reduce neuroinflammation, correlating with worsened motor function and cognitive abilities. Thus, CCI-induced behavioral deficits were also ameliorated by the administration of allicin via a PKC-δ-related mitophagy. CONCLUSIONS This study uncovers a novel mechanism where allicin enhances PKC-δ expression and PLS3 phosphorylation, facilitating CL translocation to the OMM and activating mitophagy, thereby reducing TBI-induced neuroinflammation.
Collapse
Affiliation(s)
- Yue Cheng
- Department of Radiology, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Liangxi District, Wuxi 214001, China
| | - Wei Gu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuechao Wu
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Liangxi District, Wuxi 214001, China
| | - Wei Tian
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Liangxi District, Wuxi 214001, China
| | - Zhenqian Mu
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Liangxi District, Wuxi 214001, China
| | - Yangfan Ye
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Honglu Chao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongyuan Bao
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Liangxi District, Wuxi 214001, China.
| |
Collapse
|
20
|
Chen Y, Han L, Zhu DS, Guan YT. Fibrinogen and Neuroinflammation in the Neurovascular Unit in Stroke. J Inflamm Res 2025; 18:4567-4584. [PMID: 40191094 PMCID: PMC11971976 DOI: 10.2147/jir.s496433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/13/2025] [Indexed: 04/09/2025] Open
Abstract
Stroke remains a leading cause of death and disability worldwide. Recent evidence suggests that stroke pathophysiology extends beyond vascular dysfunction to include complex interactions within the neurovascular unit (NVU), particularly involving fibrinogen. This blood-derived protein accumulates in the brain following blood-brain barrier (BBB) disruption and plays crucial roles in neuroinflammation and tissue repair. Through its unique structural domains, fibrinogen interacts with multiple cellular components, including astrocytes, microglia, and neural stem cells, thereby modulating inflammatory responses and neural repair mechanisms. This review examines fibrinogen's structure and its diverse functions in stroke pathophysiology, focusing on its interactions with vascular cells, glial cells, and peripheral immune cells. We also discuss emerging therapeutic strategies targeting fibrinogen-mediated pathways and the challenge of translating experimental results into effective clinical treatments.
Collapse
Affiliation(s)
- Yi Chen
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Lu Han
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - De-Sheng Zhu
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Department of Neurology, Baoshan Branch, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, People’s Republic of China
| | - Yang-Tai Guan
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Department of Neurology, Punan Hospital, Shanghai, 200125, People’s Republic of China
| |
Collapse
|
21
|
Gao Y, Zhang M, Wang G, Lai W, Liao S, Chen Y, Ning Q, Tang S. Metabolic cross-talk between glioblastoma and glioblastoma-associated microglia/macrophages: From basic insights to therapeutic strategies. Crit Rev Oncol Hematol 2025; 208:104649. [PMID: 39922398 DOI: 10.1016/j.critrevonc.2025.104649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Glioblastoma (GBM), a highly malignant "cold" tumor of the central nervous system, is characterized by its ability to remodel the GBM immune microenvironment (GME), leading to significant resistance to immunotherapy. GBM-associated microglia/macrophages (GAMs) are essential components of the GME. Targeting GAMs has emerged as a promising strategy against GBM. However, their highly immunosuppressive nature contributes to GBM progression and drug resistance, significantly impeding anti-GBM immunotherapy. Accumulating evidence suggests that metabolic reprogramming accompanies GBM progression and GAM polarization, which are in turn driven by specific metabolic abnormalities and altered cellular signaling pathways. Importantly, metabolic crosstalk between GBM and GAMs further promotes tumor progression. Clarifying and disrupting this metabolic crosstalk is expected to enhance the antitumor phenotype of GAMs and inhibit GBM malignant progression. This review explores metabolism-based interregulation between GBM and GAMs and summarizes recent therapeutic strategies targeting this crosstalk, offering new insights into GBM immunotherapy.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Mengxia Zhang
- Department of Histology and Embryology, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Guihua Wang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Weiwei Lai
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Shuxian Liao
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Yao Chen
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Shengsong Tang
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
22
|
Elbadawy NN, Saad MA, Elfarrash S, Ahmed MAE, Abdelkader NF. The GLP-1 agonist semaglutide ameliorates cognitive regression in P301S tauopathy mice model via autophagy/ACE2/SIRT1/FOXO1-Mediated Microglia Polarization. Eur J Pharmacol 2025; 991:177305. [PMID: 39875022 DOI: 10.1016/j.ejphar.2025.177305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Tau hyper-phosphorylation has been recognized as an essential contributor to neurodegeneration in Alzheimer's disease (AD) and related tauopathies. In the last decade, tau hyper-phosphorylation has gained considerable concern in AD therapeutic development. Tauopathies are manifested with a broad spectrum of symptoms, from dementia to cognitive decline and motor impairments. Tau undergoes conformational changes and abnormal phosphorylation that mediate its detaching from microtubules, forming neurofibrillary tangles (NFTs). In the current study, a widely used P301S transgenic mice model of tauopathy was employed to evaluate the possible neuroprotective effects of semaglutide as an autophagy regulator through modifications of the brain renin-angiotensin system (RAS). Mice were divided into two groups according to their genotypes (wild type (Wt) and P301S), which were further subdivided to receive either vehicle (saline) or semaglutide (25 nmol/kg, i. p.), once every 2 days for 28 days. Current data suggest that semaglutide ameliorated the hyperactive pattern and alleviated the cognitive decline of P301S mice. It also hastened the autophagic flux through augmenting angiotensin-converting enzyme 2/sirtuin 1/forkhead box protein O1 signaling. Semaglutide also hindered the expression of phosphorylated adenosine monophosphate-activated protein kinase and phosphorylated glycogen synthase kinase-3 beta at serine 9, reducing the propagation of neuroinflammatory cytokines and oxidative reactions. Finally, semaglutide protected against hippocampal degeneration and reduced the immunoreactivity for total tau and ionized calcium-binding adapter molecule. Semaglutide showed promising neuroprotective implications in alleviating tauopathy-related AD's molecular and behavioral deficits through controlling autophagy and brain RAS.
Collapse
Affiliation(s)
- Norhan N Elbadawy
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 12566, 6th of October City, Giza, Egypt.
| | - Muhammed A Saad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, 4184, Ajman, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Sara Elfarrash
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, 35516, Mansoura, Egypt; Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, 35516, Mansoura, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 12566, 6th of October City, Giza, Egypt
| | - Noha F Abdelkader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| |
Collapse
|
23
|
Zhang X, Cui X, Sun N, Wu X, Pan X, Wang R, Chen Z, Li Y, Hu Y, Liu F, Cao X. Microglial repopulation alleviates surgery-induced neuroinflammation and cognitive impairment in a ZEB1-dependent manner. FASEB J 2025; 39:e70440. [PMID: 40052833 DOI: 10.1096/fj.202402492r] [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: 10/16/2024] [Revised: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Microglia play a crucial role in postoperative cognitive dysfunction (POCD). This study investigated the effects of microglial depletion and subsequent repopulation on POCD and its underlying mechanisms. An aged mouse model of POCD was induced by partial hepatectomy, and the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 was administered to facilitate microglial depletion and repopulation. Neutrophil involvement was assessed with anti-Ly6G antibodies, while ZEB1 was manipulated through shRNA knockdown and lentiviral overexpression in the BV2 microglial cell line. A TGF-β1 neutralizing antibody was employed to elucidate the relationship between ZEB1 and its downstream pathways. The results indicated that microglial depletion alone did not reverse cognitive impairments. However, microglial repopulation significantly reduced neutrophil infiltration and improved cognitive function post-surgery. This improvement correlated with ZEB1 upregulation in microglia, which decreased CXCL1 production by astrocytes via TGF-β1 signaling, thereby reducing neutrophil migration to the hippocampus. These findings suggest that microglial repopulation, dependent on ZEB1 and TGF-β1 signaling, effectively alleviates neuroinflammation, reduces neutrophil infiltration, and enhances cognitive function, highlighting microglia as a promising target for the prevention and treatment of POCD.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaotong Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Naihui Sun
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinyi Wu
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xue Pan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renyi Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zitong Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yilong Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Hu
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fang Liu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuezhao Cao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Li X, Kim HJ, Yoo J, Lee Y, Nam CH, Park J, Lee ST, Kim TM, Choi SH, Won JK, Park SH, Ju YS, Park JB, Kim SH, Chang JH, Wu HG, Park CK, Lee JH, Kang SG, Lee JH. Distant origin of glioblastoma recurrence: neural stem cells in the subventricular zone serve as a source of tumor reconstruction after primary resection. Mol Cancer 2025; 24:64. [PMID: 40033380 PMCID: PMC11877783 DOI: 10.1186/s12943-025-02273-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/15/2025] [Indexed: 03/05/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive and common type of primary malignant brain cancer in adults. GBM often recurs locally near the resection cavity (RC) following the surgical removal of primary tumors. Recent research has reported that neural stem cells (NSCs) in the subventricular zone (SVZ) harboring cancer-driving mutations serve as the cells of origin for human GBM. However, the pathological role of tumor-initiating NSCs in the SVZ in tumor recurrence remains to be elucidated. Here, we explore the potential contribution of mutation-harboring NSCs in the SVZ to tumor recurrence around the RC following surgical resection. Our hypothesis emerged from performing deep sequencing of longitudinal tissues from 10 patients with GBM, including (i) tumor-free SVZ tissue, (ii) primary tumor tissue, (iii) recurrent tumor tissue, and (iv) blood. As a result of this sequencing, we observed evidence suggesting that recurrent tumors show genetic links to the SVZ in 60% (6/10) of patients, which are distinct from the primary tumors. Using a genome-edited mouse model, we further identified that mutation-harboring NSCs appeared to migrate to the RC through the aberrant growth of oligodendrocyte progenitor cells, potentially contributing to the reconstruction of high-grade malignant gliomas in the RC. This process was associated with the CXCR4/CXCL12 axis, as supported by RNA sequencing data from human recurrent GBM. Taken together, our findings suggest that NSCs in human SVZ tissue may play a role in GBM recurrence, potentially highlighting a novel distant contributor of recurrence.
Collapse
Affiliation(s)
- Xue Li
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Hyun Jung Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Jihwan Yoo
- Department of Neurosurgery, Brain Tumor Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Yeonhee Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Chang Hyun Nam
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jonghan Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jong Bae Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hong-Gyun Wu
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
- Sovargen Inc, Daejeon, South Korea.
| | - Seok-Gu Kang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.
- Department of Medical Sciences, Yonsei University Graduate School, Seoul, South Korea.
| | - Joo Ho Lee
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
25
|
Yu L, Sun L, Yu T, Guo A, Wu J, Chen J, Wang Q. CPCGI Alleviates Neural Damage by Modulating Microglial Pyroptosis After Traumatic Brain Injury. CNS Neurosci Ther 2025; 31:e70322. [PMID: 40059065 PMCID: PMC11890976 DOI: 10.1111/cns.70322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major global cause of mortality and long-term disability, with limited therapeutic options. Microglial pyroptosis, a form of programmed cell death associated with inflammation, has been implicated in exacerbating neuroinflammation and secondary injury following TBI. Compound porcine cerebroside ganglioside injection (CPCGI) has shown anti-inflammatory and antioxidant properties, but its effects on pyroptosis remain unexplored. This study investigates the role of CPCGI in TBI and its underlying mechanisms. METHODS A controlled cortical impact (CCI) model was utilized to establish TBI in vivo, while lipopolysaccharide (LPS) was used in vitro to induce microglial activation that mimicked TBI conditions. The effects of CPCGI on microglial pyroptosis and inflammatory cytokines were analyzed through immunofluorescence, flow cytometry, western blotting, and quantitative real-time PCR (qRT-PCR). The involvement of the NLRP3 inflammasome in CPCGI's mechanism was examined using NLRP3 overexpression or the NLRP3 agonist BMS-986299. A microglia-neuron interaction model was created, and neuronal injury was assessed with the Cell Counting Kit-8 and Fluoro-Jade C (FJC). RESULTS Treatment with CPCGI resulted in significant improvement in the neurobehavioral outcomes, reduced lesion volume, and decreased neuronal loss following TBI. Notably, TBI induced microglial pyroptosis and the release of pro-inflammatory cytokines, while CPCGI inhibited microglial pyroptosis, thereby mitigating the inflammatory response and reducing neuronal damage. Mechanistically, overexpression of NLRP3 in microglial cells reversed the inhibitory effects of CPCGI on microglial pyroptosis, indicating that CPCGI's inhibition of microglial pyroptosis may be mediated by the NLRP3 inflammasome. Furthermore, NLRP3 overexpression or administration of the NLRP3 agonist BMS-986299 negated the neuroprotective effects of CPCGI in vivo and in vitro. CONCLUSION These findings suggest that CPCGI provides neuroprotection in TBI by targeting NLRP3 inflammasome-mediated microglial pyroptosis, thereby improving the neuroinflammatory microenvironment and promoting neurological recovery. This underscores its potential as a promising candidate for TBI treatment.
Collapse
Affiliation(s)
- Lu‐Lu Yu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Lei Sun
- Department of NeurologyZhengzhou University People's HospitalZhengzhouChina
| | - Ting‐Ting Yu
- Department of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - An‐Chen Guo
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Drug and Device Research and Development for Cerebrovascular DiseasesBeijingChina
| | - Jian‐Ping Wu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Key Laboratory of Drug and Device Research and Development for Cerebrovascular DiseasesBeijingChina
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingChina
| | - Jun‐Min Chen
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qun Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
26
|
Afridi R, Bhusal A, Lee SE, Hwang EM, Ryu H, Kim JH, Suk K. A microglial kinase ITK mediating neuroinflammation and behavioral deficits in traumatic brain injury. Mol Cell Neurosci 2025; 132:103994. [PMID: 39864680 DOI: 10.1016/j.mcn.2025.103994] [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: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025] Open
Abstract
Microglia-mediated neuroinflammation has been implicated in the neuropathology of traumatic brain injuries (TBI). Recently, the expression of interleukin-2-inducible T-cell kinase (ITK) has been detected in brain microglia, regulating their inflammatory activities. However, the role of microglial ITK in TBI has not been investigated. In this study, we demonstrate that ITK expression and activation are upregulated in microglia following an injury caused by controlled cortical impact (CCI) - a mouse model of TBI. Pharmacological inhibition of ITK protein or knockdown of microglial ITK gene expression using adeno-associated virus mitigates neuroinflammation and improves neurological outcomes in the CCI model. Additionally, ITK mRNA expression was found to be increased in the brains of patients with chronic traumatic encephalopathy. An ITK inhibitor reduced the activation of inflammatory responses in both human and mouse microglia in vitro. Collectively, these results suggest that microglial ITK plays a pivotal role in neuroinflammation and mediating behavioral deficits following TBI. Thus, targeting the signaling pathway of microglial ITK may exert protective effects by alleviating neuroinflammation associated with TBI.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Anup Bhusal
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jong-Heon Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
27
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial Autophagic Dysregulation in Traumatic Brain Injury: Molecular Insights and Therapeutic Avenues. ACS Chem Neurosci 2025; 16:543-562. [PMID: 39920904 DOI: 10.1021/acschemneuro.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted condition that can result in cognitive and behavioral impairments. One aspect of TBI that has received increasing attention in recent years is the role of microglia, the brain-resident immune cells, in the pathophysiology of the injury. Specifically, increasing evidence suggests that dysfunction in microglial autophagy, the process by which cells degrade and recycle their own damaged components, may contribute to the development and progression of TBI-related impairments. Here, we unravel the pathways by which microglia autophagic dysregulation predisposes the brain to secondary damage and neurological deficits following TBI. An overview of the role of autophagic dysregulation in perpetuation and worsening of the inflammatory response, neuroinflammation, and neuronal cell death in TBI follows. Further, we have evaluated several signaling pathways and processes that contribute to autophagy dysfunction-mediated inflammation, neurodegeneration, and poor outcome in TBI. Additionally, a discussion on the small molecule therapeutics employed to modulate these pathways and mechanisms to treat TBI have been presented. However, additional research is required to fully understand the processes behind these underlying pathways and uncover potential therapeutic targets for restoring microglial autophagic failure in TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, California 92037, United States
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University, Sakaka 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research, New Delhi 201002, India
| | | |
Collapse
|
28
|
Packer JM, Giammo SG, Wangler LM, Davis AC, Bray CE, Godbout JP. Diffuse Traumatic Brain Injury Induced Stimulator of Interferons (STING) Signaling in Microglia Drives Cortical Neuroinflammation, Neuronal Dysfunction, and Impaired Cognition. RESEARCH SQUARE 2025:rs.3.rs-5960640. [PMID: 40034431 PMCID: PMC11875282 DOI: 10.21203/rs.3.rs-5960640/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: 03/05/2025]
Abstract
Neuropsychiatric complications including depression and cognitive impairment develop, persist, and worsen in the years after traumatic brain injury (TBI), negatively affecting life and lifespan. Inflammatory responses mediated by microglia are associated with the transition from acute to chronic neuroinflammation after TBI. Moreover, type I interferon (IFN-I) signaling is a key mediator of inflammation during this transition. Thus, the purpose of this study was to determine the degree to which a microglia-specific knockout of the stimulator of interferons (STING) influenced TBI-induced neuroinflammation, neuronal dysfunction, and cognitive impairment. Here, microglial inducible STING knockout (CX3CR1Cre/ERT2 × STINGfl/fl) mice were created and validated (mSTING-/-). Diffuse brain injury (midline fluid percussion) in male and female mice increased STING expression in microglia, promoted microglial morphological restructuring, and induced robust cortical inflammation and pathology 7 days post injury (dpi). These TBI-associated responses were attenuated in mSTING-/- mice. Increased cortical astrogliosis and rod-shaped microglia induced by TBI were independent of mSTING-/-. 7 dpi, TBI induced 237 differentially expressed genes (DEG) in the cortex of functionally wildtype (STING+/+) associated with STING, NF-κB, and Interferon Alpha signaling and 85% were attenuated by mSTING-/-. Components of neuronal injury including reduced NeuN expression, increased cortical lipofuscin, and increased neurofilament light chain in plasma were increased by TBI and dependent on mSTING. TBI-associated cognitive tasks (novel object recognition/location, NOR/NOL) at 7 dpi were dependent on mSTING. Notably, the TBI-induced cognitive deflcits in NOR/NOL and increased cortical inflammation 7 dpi were unaffected in global interferon-α/β receptor 1 knockout (IFNAR1) mice. In the final study, the RNA profile of neurons after TBI in STING+/+ and mSTING-/- mice was assessed 7 dpi by single nucleus RNA-sequencing. There was a TBI-dependent suppression of cortical neuronal homeostasis with reductions in CREB signaling, synaptogenesis, and oxytocin signaling and increases in cilium assembly and PTEN signaling. Overall, mSTING-/- prevented 50% of TBI-induced DEGs in cortical neurons. Collectively, ablation of STING in microglia attenuates TBI-induced IFN-dependent responses, cortical inflammation, neuronal dysfunction, neuronal pathology, and cognitive impairment.
Collapse
|
29
|
Chen Z, Zhang C, Fang Y, Zhang H, Luo J, Miao C, Li J, Peng J, Qiu Y, Xia Y, Luo Q. Olfactory mucosa-mesenchymal stem cells with overexpressed Nrf2 modulate angiogenesis and exert anti-inflammation effect in an in vitro traumatic brain injury model. Eur J Med Res 2025; 30:80. [PMID: 39910594 PMCID: PMC11796021 DOI: 10.1186/s40001-025-02344-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major cause of disability and mortality among children and adults in developed countries. Transcription factor nuclear factor erythroid-derived 2-like 2 (Nrf2) has antioxidant, anti-inflammatory and neuroprotective effects and is closely related to TBI. Olfactory mucosa-mesenchymal stem cells (OM-MSCs) could promote neural regeneration. At present, the effects of OM-MSCs with overexpressed Nrf2 in brain diseases remain to be explored. METHODS The OM-MSCs were prepared and transfected with Nrf2 overexpression plasmid. Those transfected cells were termed as OM-MSCs with Nrf2 overexpression (OM-MSCsNrf2) and co-cultured with rat pheochromocytoma cells PC12 or murine microglia BV2. The effects of OM-MSCsNrf2 on the survival and angiogenesis of PC12 cells were evaluated through cell counting kit-8 (CCK-8) and tube formation assay, and extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) were calculated to reflect glycolysis. Immunofluorescence assay was applied to determine the effects of OM-MSCsNrf2 on microglial polarization, and the underlying molecular mechanisms were analyzed based on the quantification tests of RT-qPCR and immunoblotting. RESULTS Co-culture of OM-MSCsNrf2 and PC12 cells increased the levels of anti-inflammatory cytokines and pro-angiogenesis factors, enhanced the cell survival and angiogenesis. Moreover, we also observed elevated phosphorylation of PI3K/AKT and suppressed BAX protein expression. Meanwhile, OM-MSCsNrf2 inhibited the levels of pro-inflammatory genes and affected the glycolysis in PC12 cells. In the co-cultured system of OM-MSCsNrf2 and BV2 cells, M2 microglial polarization was observed, and the levels of M2 microglia-relevant genes and the phosphorylation of STAT6/AMPKα/SMAD3 were elevated. CONCLUSION This study proved the effects of OM-MSCsNrf2 on modulating PC12 and BV2 cells in vitro, which, however, necessitates further in vivo validation.
Collapse
Affiliation(s)
- Zigui Chen
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Chunyuan Zhang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, 533000, China
| | - Yuhua Fang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, 533000, China
| | - He Zhang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, 533000, China
| | - Jiawei Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, 533000, China
| | - Changfeng Miao
- Department of Neurosurgery Second Branche, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Jiale Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Yingqi Qiu
- Department of Clinical Research Center, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| | - Qisheng Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, 533000, China.
| |
Collapse
|
30
|
Biltz RG, Yin W, Goodman EJ, Wangler LM, Davis AC, Oliver BT, Godbout JP, Sheridan JF. Repeated social defeat in male mice induced unique RNA profiles in projection neurons from the amygdala to the hippocampus. Brain Behav Immun Health 2025; 43:100908. [PMID: 39720627 PMCID: PMC11667635 DOI: 10.1016/j.bbih.2024.100908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Chronic stress increases the incidence of psychiatric disorders including anxiety, depression, and posttraumatic stress disorder. Repeated Social Defeat (RSD) in mice recapitulates several key physiological, immune, and behavioral changes evident after chronic stress in humans. For instance, neurons in the prefrontal cortex, amygdala, and hippocampus are involved in the interpretation of and response to fear and threatful stimuli after RSD. Therefore, the purpose of this study was to determine how stress influenced the RNA profile of hippocampal neurons and neurons that project into the hippocampus from threat appraisal centers. Here, RSD increased anxiety-like behavior in the elevated plus maze and reduced hippocampal-dependent novel object location memory in male mice. Next, pan-neuronal (Baf53 b-Cre) RiboTag mice were generated to capture ribosomal bound mRNA (i.e., active translation) activated by RSD in the hippocampus. RNAseq revealed that there were 1694 differentially expressed genes (DEGs) in hippocampal neurons after RSD. These DEGs were associated with an increase in oxidative stress, synaptic long-term potentiation, and neuroinflammatory signaling. To further examine region-specific neural circuitry associated with fear and anxiety, a retrograde-adeno-associated-virus (AAV2rg) expressing Cre-recombinase was injected into the hippocampus of male RiboTag mice. This induced expression of a hemagglutinin epitope in neurons that project into the hippocampus. These AAV2rg-RiboTag mice were subjected to RSD and ribosomal-bound mRNA was collected from the amygdala for RNA-sequencing. RSD induced 677 DEGs from amygdala projections. Amygdala neurons that project into the hippocampus had RNA profiles associated with increased synaptogenesis, interleukin-1 signaling, nitric oxide, and reactive oxygen species production. Using a similar approach, there were 1132 DEGs in neurons that project from the prefrontal cortex. These prefrontal cortex neurons had RNA profiles associated with increased synaptogenesis, integrin signaling, and dopamine feedback signaling after RSD. Collectively, there were unique RNA profiles of stress-influenced projection neurons and these profiles were associated with hippocampal-dependent behavioral and cognitive deficits.
Collapse
Affiliation(s)
- Rebecca G. Biltz
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
| | - Wenyuan Yin
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
| | - Ethan J. Goodman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
| | - Lynde M. Wangler
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
| | - Amara C. Davis
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
| | - Braedan T. Oliver
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, USA
| | - Jonathan P. Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
- Chronic Brain Injury Program, The Ohio State University, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, USA
| | - John F. Sheridan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, USA
- Division of Biosciences, The Ohio State University College of Dentistry, USA
- Chronic Brain Injury Program, The Ohio State University, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, USA
| |
Collapse
|
31
|
Tak H, Anirudh J, Chattopadhyay A, Naick BH. Argonaute protein assisted drug discovery for miRNA-181c-5p and target gene ATM translation repression: a computational approach. Mol Divers 2025; 29:351-365. [PMID: 39026118 DOI: 10.1007/s11030-024-10855-3] [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: 12/26/2023] [Accepted: 03/21/2024] [Indexed: 07/20/2024]
Abstract
The miRNA binds to AGO's seed region, prompting the exploration of small molecules that can offset miRNA repression of target mRNA. This miRNA-181c-5p was found to be upregulated in the chronic traumatic encephalopathy, a prevalent neurodegenerative disease in contact sports and military personals. The research aimed to identify compounds that disrupt the AGO-assisted loop formation between miRNA-181c-5p and ATM, consequently repressing the translation of ATM. Target genes from commonly three databases (DIANA-microT-CDS, miRDB, RNA22 and TargetScan) were subjected to functional annotation and clustering analysis using DAVID bioinformatics tool. Haddock server were employed to make miRNA-181c-5p:ATM-AGO complex. A total of 2594 small molecules were screened using Glide XP based on their highest binding affinity towards the complex, through a three-phase docking approach. The top 5 compounds (DB00674-Galantamine, DB00371-Meprobamate, DB00694-Daunorubicin, DB00837-Progabide, and DB00851-Dacarbazine) were further analyzed for stability in the miRNA-181c-5p:ATM-AGO-ligand complex interaction using GROMACS (version 2023.2). Hence, these findings suggest that these molecules hold potential for facilitating AGO-assisted repression of ATM gene translation.
Collapse
Affiliation(s)
- Harshita Tak
- Department of Sports Biosciences, School of Sports Science, Central University of Rajasthan, Ajmer, India
| | - Jivanage Anirudh
- Department of Sports Biosciences, School of Sports Science, Central University of Rajasthan, Ajmer, India
| | - Arpan Chattopadhyay
- Department of Sports Biosciences, School of Sports Science, Central University of Rajasthan, Ajmer, India
| | - B Hemanth Naick
- Department of Sports Biosciences, School of Sports Science, Central University of Rajasthan, Ajmer, India.
| |
Collapse
|
32
|
Ye G, Wang Z, Chen P, Ye J, Li S, Chen M, Feng J, Wang H, Chen W. Serpina3n in neonatal microglia mediates its protective role for damaged adult microglia by alleviating extracellular matrix remodeling-induced tunneling nanotubes degradation in a cell model of traumatic brain injury. Neuroscience 2025; 565:1-9. [PMID: 39613247 DOI: 10.1016/j.neuroscience.2024.11.066] [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: 07/05/2024] [Revised: 10/26/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Traumatic brain injury (TBI) induces significant neuroinflammation, primarily driven by microglia. Neonatal microglia (NMG) may have therapeutic potential by modulating the inflammatory response of damaged adult microglia (AMG). This study investigates the influence of NMG on AMG function through extracellular matrix (ECM) remodeling and the formation of tunneling nanotubes (TnTs), with a focus on the role of Serpina3n. We established an in vitro TBI model using a 3D Transwell system, co-culturing damaged AMG with NMG. Viral vector transfection was employed to manipulate Serpina3n expression in NMG. Quantitative real-time PCR, Western blotting, and ELISA were utilized to assess inflammatory markers, ECM remodeling proteins, and TnTs-related proteins. Co-culturing with NMG significantly inhibited M1 polarization of AMG and reduced the release of pro-inflammatory cytokines while promoting M2 polarization and increasing the production of anti-inflammatory cytokines. NMG expressed higher levels of Serpina3n, which played a crucial role in reducing Granzyme B, matrix metalloproteinase (MMP) 2 and MMP9 expression, thereby mitigating ECM remodeling. Inhibition of Serpina3n in NMG increased pro-inflammatory markers and decreased TnTs formation proteins, whereas overexpression of M-sec in AMG counteracted these effects. This highlights the importance of TnTs in maintaining microglial function and promoting an anti-inflammatory environment. In conclusion, NMG improve the function of damaged AMG by modulating ECM remodeling and promoting TnTs formation through the action of Serpina3n.
Collapse
Affiliation(s)
- Gengfan Ye
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China
| | - Zhigang Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Pudong New Area, Shanghai 200120, China
| | - Pandi Chen
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China
| | - Junyi Ye
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China
| | - Shiwei Li
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China
| | - Maosong Chen
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China
| | - Jiugeng Feng
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Hongcai Wang
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China
| | - Wei Chen
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University. Ningbo 315040, Zhejiang, China; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Pudong New Area, Shanghai 200120, China.
| |
Collapse
|
33
|
Feichtenbiner AB, Sytsma K, O'Boyle RP, Mittenzwei R, Maioli H, Scherpelz KP, Child DD, Li N, Ariza Torres J, Keene L, Kirkland A, Howard K, Latimer C, Keene CD, Ransom C, Nolan AL. Satellite microglia: marker of traumatic brain injury and regulator of neuronal excitability. J Neuroinflammation 2025; 22:9. [PMID: 39819341 PMCID: PMC11740464 DOI: 10.1186/s12974-024-03328-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
Traumatic brain injury is a leading cause of chronic neurologic disability and a risk factor for development of neurodegenerative disease. However, little is known regarding the pathophysiology of human traumatic brain injury, especially in the window after acute injury and the later life development of progressive neurodegenerative disease. Given the proposed mechanisms of toxic protein production and neuroinflammation as possible initiators or contributors to progressive pathology, we examined phosphorylated tau accumulation, microgliosis and astrogliosis using immunostaining in the orbitofrontal cortex, a region often vulnerable across traumatic brain injury exposures, in an age and sex-matched cohort of community traumatic brain injury including both mild and severe cases in midlife. We found that microglial response is most prominent after chronic traumatic brain injury, and interactions with neurons in the form of satellite microglia are increased, even after mild traumatic brain injury. Taking our investigation into a mouse model, we identified that these satellite microglia suppress neuronal excitability in control conditions but lose this ability with chronic traumatic brain injury. At the same time, network hyperexcitability is present in both mouse and human orbitofrontal cortex. Our findings support a role for loss of homeostatic control by satellite microglia in the maladaptive circuit changes that occur after traumatic brain injury.
Collapse
Affiliation(s)
- Alicia B Feichtenbiner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Karinn Sytsma
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Ryan P O'Boyle
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Rhonda Mittenzwei
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
- King County Office of the Medical Examiner, Seattle, WA, 98104, USA
| | - Heather Maioli
- Office of Chief Medical Examiner of the City of New York, New York, NY, 10016, USA
| | - Kathryn P Scherpelz
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Daniel D Child
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Ning Li
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA
| | | | - Lisa Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Amanda Kirkland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Kimberly Howard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Caitlin Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Christopher Ransom
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA.
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA.
| |
Collapse
|
34
|
Yang X, Tang C, Zhu D, Xia X, Du Q, Huang L, Liu J, Liu Y. Nonylphenol exposure increases the risk of Hirschsprung's disease by inducing macrophage M1 polarization. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117756. [PMID: 39837008 DOI: 10.1016/j.ecoenv.2025.117756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Nonylphenol (NP), a ubiquitous environmental contaminant used as a surfactant in industrial production and classified as an endocrine disruptor, could interfere hormone secretion and exhibit neurotoxicity in organisms. Hirschsprung's disease (HSCR), one of the most frequently observed congenital malformations of the digestive system, arises mainly due to the failure of enteric neural crest cells to migrate to the distal colon during embryonic development. However, the effects of NP exposure on HSCR are largely unknown. Herein, we identified the content of NP and expression of lncRNA LINC00294/Inhibin Subunit Beta E (INHBE) axis in clinical samples and evaluated the crucial role of lncRNA LINC00294/INHBE axis in the neurogenic potential of neurons and the neurotoxicity effects of NP in the SH-SY5Y cells and female specific pathogen-free (SPF) rat model. Our results showed that NP concentration and LINC00294 were significantly associated with HSCR occurrence and macrophage polarization in human HSCR specimens. Moreover, NP promoted macrophage M1 polarization. The proliferation and migration were weakened, and apoptosis was heightened by the conditioned medium of NP-treated macrophages in SH-SY5Y cells. Contrarily, LINC00294 overexpression and INHBE knockdown reversed the neurotoxicity effect of NP on SH-SY5Y cells. Furthermore, the neurotoxicity effect of NP was abolished by clodronate liposomes in the rat model. In conclusion, NP could induce macrophage M1 polarization via the LINC00294/INHBE axis and increase the risk of Hirschsprung's disease. Our findings would provide a foundation for the toxicity study and risk assessments of NP.
Collapse
Affiliation(s)
- Xuefeng Yang
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China; Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chengyan Tang
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Daiwei Zhu
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Xingrong Xia
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Qing Du
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Lu Huang
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Jianguo Liu
- Key Laboratory of Oral Disease Research, School of Stomatology, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Yuanmei Liu
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China.
| |
Collapse
|
35
|
Wang L, Ouyang D, Li L, Cao Y, Wang Y, Gu N, Zhang Z, Li Z, Tang S, Tang H, Zhang Y, Sun X, Yan J. TREM2 affects DAM-like cell transformation in the acute phase of TBI in mice by regulating microglial glycolysis. J Neuroinflammation 2025; 22:6. [PMID: 39800730 PMCID: PMC11727224 DOI: 10.1186/s12974-025-03337-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is characterized by high mortality and disability rates. Disease-associated microglia (DAM) are a newly discovered subtype of microglia. However, their presence and function in the acute phase of TBI remain unclear. Although glycolysis is important for microglial differentiation, its regulatory role in DAM transformation during the acute phase of TBI is still unclear. In this study, we investigated the functions of DAM-like cells in the acute phase of TBI in mice, as well as the relationship between their transformation and glycolysis. METHODS In this study, a controlled cortical impact model was used to induce TBI in adult male wild-type (WT) C57BL/6 mice and adult male TREM2 knockout mice. Various techniques were used to assess the role of DAM-like cells in TBI and the effects of glycolysis on DAM-like cells, including RT‒qPCR, immunofluorescence assays, behavioural tests, extracellular acidification rate (ECAR) tests, Western blot analysis, cell magnetic sorting and culture, glucose and lactate assays, and flow cytometry. RESULTS DAM-like cells were observed in the acute phase of TBI in mice, and their transformation depended on TREM2 expression. TREM2 knockout impaired neurological recovery in TBI mice, possibly due in part to their role in clearing debris and secreting VEGFa and BDNF. Moreover, DAM-like cells exhibited significantly increased glycolytic activity. TREM2 regulated the AKT‒mTOR‒HIF-1α pathway and glycolysis in microglia in the acute phase of TBI. The increase in glycolysis in microglia partially contributed to the transformation of DAM-like cells in the acute phase of TBI in mice. CONCLUSIONS Taken together, the results of our study demonstrated that DAM-like cells were present in the acute phase of TBI in mice. TREM2 might influence DAM-like cell transformation by modulating the glycolysis of microglia. Our results provide a new possible pathway for intervening TBI.
Collapse
Affiliation(s)
- Lin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurosurgery, The Second Clinical Medical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, China
| | - Diqing Ouyang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lin Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yunchuan Cao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yingwen Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Nina Gu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhaosi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhao Li
- Emergency Department, Chengdu First People's Hospital, Chengdu, 610000, China
| | - Shuang Tang
- Department of Neurosurgery, Suining Central Hospital, Suining, 629000, China
| | - Hui Tang
- Department of Neurosurgery, The Second Clinical Medical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, China
| | - Yuan Zhang
- Department of Neurosurgery, The Second Clinical Medical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, China.
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Jin Yan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
36
|
Henry RJ, Loane DJ. Unraveling the complexity of microglial responses in traumatic brain and spinal cord injury. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:113-132. [PMID: 40148040 DOI: 10.1016/b978-0-443-19102-2.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Microglia, the resident innate immune cells of the central nervous system (CNS), play an important role in neuroimmune signaling, neuroprotection, and neuroinflammation. In the healthy CNS, microglia adopt a surveillant and antiinflammatory phenotype characterized by a ramified scanning morphology that maintains CNS homeostasis. In response to acquired insults, such as traumatic brain injury (TBI) or spinal cord injury (SCI), microglia undergo a dramatic morphologic and functional switch to that of a reactive state. This microglial switch is initially protective and supports the return of the injured tissue to a physiologic homeostatic state. However, there is now a significant body of evidence that both TBI and SCI can result in a chronic state of microglial activation, which contributes to neurodegeneration and impairments in long-term neurologic outcomes in humans and animal models. In this review, we discuss the complex role of microglia in the pathophysiology of TBI and SCI, and recent advancements in knowledge of microglial phenotypic states in the injured CNS. Furthermore, we highlight novel therapeutic strategies targeting chronic microglial responses in experimental models and discuss how they may ultimately be translated to the clinic for human brain and SCI.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Pharmacology, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Brieck KA, Brieck ZJ, Ashby JA, Phelps OC, Cernak I. A Narrative Review of the Effects of Internal Jugular Vein Compression on Brain Structure and Function During Periods of Head Impact. Cureus 2025; 17:e77625. [PMID: 39834663 PMCID: PMC11743508 DOI: 10.7759/cureus.77625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
Subconcussive impacts are very common in the sports world and can have many negative impacts on human function, including increased risk for cognitive decline and behavioral impairments such as chronic traumatic encephalopathy (CTE). The purpose of this article is to analyze the available literature on the effects of jugular vein compression applied by a cervical collar on cerebral structure and function in the setting of chronic impact exposure. This narrative review analyzed 17 articles on brain structure and function, published between 1992 and 2022. Our review of the 17 studies shows an overall neuroprotective effect of the external jugular vein compression applied by the cervical collar during insult to the head as compared to groups who did not wear a collar. These findings suggest a potential role of the cervical collar, in addition to helmets, in reducing the incidence of concussion-induced microtraumas and cascading secondary injury mechanisms. Though positive results are consistent throughout the studies, future studies with increased sample sizes are necessary to create precise estimates of the effects of the cervical collar. In addition, the analyzed studies mainly looked at the effects of the cervical collar on football players, soccer players, and Special Weapons and Tactics (SWAT) team members; thus, additional rigorous studies are needed to assess the impact of the cervical collar on other high-risk populations such as military and law-enforcement personnel, among others.
Collapse
Affiliation(s)
- Kathryn A Brieck
- Neurology, Augusta University Medical College of Georgia, Augusta, USA
| | | | - John A Ashby
- Neurology, Mercer University School of Medicine, Macon, USA
| | - Owen C Phelps
- Neurology, Mercer University School of Medicine, Macon, USA
| | - Ibolja Cernak
- Physiology, Thomas F. Frist, Jr. College of Medicine, Nashville, USA
| |
Collapse
|
38
|
Pinto-Benito D, Bautista-Abad A, Lagunas N, Ontiveros N, Ganchala D, Garcia-Segura LM, Arevalo MA, Grassi D. Tibolone treatment after traumatic brain injury exerts a sex-specific and Y chromosome-dependent regulation of methylation and demethylation enzymes and estrogen receptors in the cerebral cortex. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167532. [PMID: 39366643 DOI: 10.1016/j.bbadis.2024.167532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Affiliation(s)
- Daniel Pinto-Benito
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alvaro Bautista-Abad
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natalia Lagunas
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, Ciudad Universitaria, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Nebai Ontiveros
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Danny Ganchala
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis M Garcia-Segura
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria-Angeles Arevalo
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Daniela Grassi
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Anatomy, Histology and Neuroscience, School of Medicine, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, 28029 Madrid, Spain
| |
Collapse
|
39
|
León-Rodríguez A, Grondona JM, Marín-Wong S, López-Aranda MF, López-Ávalos MD. Long-term reprogramming of primed microglia after moderate inhibition of CSF1R signaling. Glia 2025; 73:175-195. [PMID: 39448548 DOI: 10.1002/glia.24627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
In acute neuroinflammation, microglia activate transiently, and return to a resting state later on. However, they may retain immune memory of such event, namely priming. Primed microglia are more sensitive to new stimuli and develop exacerbated responses, representing a risk factor for neurological disorders with an inflammatory component. Strategies to control the hyperactivation of microglia are, hence, of great interest. The receptor for colony stimulating factor 1 (CSF1R), expressed in myeloid cells, is essential for microglia viability, so its blockade with specific inhibitors (e.g. PLX5622) results in significant depletion of microglial population. Interestingly, upon inhibitor withdrawal, new naïve microglia repopulate the brain. Depletion-repopulation has been proposed as a strategy to reprogram microglia. However, substantial elimination of microglia is inadvisable in human therapy. To overcome such drawback, we aimed to reprogram long-term primed microglia by CSF1R partial inhibition. Microglial priming was induced in mice by acute neuroinflammation, provoked by intracerebroventricular injection of neuraminidase. After 3-weeks recovery, low-dose PLX5622 treatment was administrated for 12 days, followed by a withdrawal period of 7 weeks. Twelve hours before euthanasia, mice received a peripheral lipopolysaccharide (LPS) immune challenge, and the subsequent microglial inflammatory response was evaluated. PLX5622 provoked a 40%-50% decrease in microglial population, but basal levels were restored 7 weeks later. In the brain regions studied, hippocampus and hypothalamus, LPS induced enhanced microgliosis and inflammatory activation in neuraminidase-injected mice, while PLX5622 treatment prevented these changes. Our results suggest that PLX5622 used at low doses reverts microglial priming and, remarkably, prevents broad microglial depletion.
Collapse
Affiliation(s)
- Ana León-Rodríguez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Sonia Marín-Wong
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Manuel F López-Aranda
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| |
Collapse
|
40
|
Liang X, Hu Y, Li X, Xu X, Chen Z, Han Y, Han Y, Lang G. Role of PI3Kγ in the polarization, migration, and phagocytosis of microglia. Neurochem Int 2025; 182:105917. [PMID: 39675432 DOI: 10.1016/j.neuint.2024.105917] [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: 09/30/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Phosphoinositide 3-kinase γ (PI3Kγ) is a signaling protein that is constitutively expressed in immune competent cells and plays a crucial role in cell proliferation, apoptosis, migration, deformation, and immunology. Several studies have shown that high expression of PI3Kγ can inhibit the occurrence of inflammation in microglia while also regulating the polarization of microglia to inhibit inflammation and enhance microglial migration and phagocytosis. It is well known that the regulation of microglial polarization, migration, and phagocytosis is key to the treatment of most neurodegenerative diseases. Therefore, in this article, we review the important regulatory role of PI3Kγ in microglia to provide a basis for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xinghua Liang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, 563000, China.
| | - Yuan Hu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, 563000, China.
| | - Xinyue Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, 563000, China.
| | - Xi Xu
- The Special Key Laboratory of Oral Diseases Research Institution of Higher Education in Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| | - Zhonglan Chen
- The Special Key Laboratory of Oral Diseases Research Institution of Higher Education in Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| | - Yalin Han
- The Special Key Laboratory of Oral Diseases Research Institution of Higher Education in Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| | - Yingying Han
- The Special Key Laboratory of Oral Diseases Research Institution of Higher Education in Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| | - Guangping Lang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
41
|
Li P, Ye L, Sun S, Wang Y, Chen Y, Chang J, Yin R, Liu X, Zuo W, Xu H, Zhang X, Zhao RC, Han Q, Wei J. Molecular intersections of traumatic brain injury and Alzheimer's disease: the role of ADMSC-derived exosomes and hub genes in microglial polarization. Metab Brain Dis 2024; 40:77. [PMID: 39715972 DOI: 10.1007/s11011-024-01503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024]
Abstract
Traumatic brain injury (TBI) is a significant contributor to global mortality and morbidity, with emerging evidence indicating a heightened risk of developing Alzheimer's disease (AD) following TBI. This study aimed to explore the molecular intersections between TBI and AD, focusing on the role of adipose mesenchymal stem cell (ADMSC)-derived exosomes and hub genes involved in microglial polarization. Transcriptome profiles from TBI (GSE58485) and AD (GSE74614) datasets were analyzed to identify differentially expressed genes (DEGs). The hub genes were validated in independent datasets (GSE180811 for TBI and GSE135999 for AD) and localized to specific cell types using single-cell RNA (scRNA) sequencing data (GSE160763 for TBI and GSE224398 for AD). Experimental validation was conducted to investigate the role of these genes in microglial polarization using cell culture and ADMSC-derived exosomes interventions. Our results identified three hub genes-Bst2, B2m, and Lgals3bp-that were upregulated in both TBI and AD, with strong associations to inflammation, neuronal apoptosis, and tissue repair processes. scRNA sequencing revealed that these genes are predominantly expressed in microglia, with increased expression during M1 polarization. Knockdown of these genes reduced M1 polarization and promoted M2 phenotype in microglia. Additionally, ADMSC-derived exosomes attenuated M1 polarization and downregulated the expression of hub genes. This study provides novel insights into the shared molecular pathways between TBI and AD, highlighting potential therapeutic targets for mitigating neuroinflammation and promoting recovery in both conditions.
Collapse
Affiliation(s)
- Pengtao Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liguo Ye
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Sishuai Sun
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yihao Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianbo Chang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Yin
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyu Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Zuo
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Houshi Xu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Junji Wei
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
42
|
Xia M, Yi M, Guo C, Xie Y, Yu W, Wang D, Dai X. β-Asarone regulates microglia polarization to alleviate TBI-induced nerve damage via Fas/FasL signaling axis. Hum Cell 2024; 38:33. [PMID: 39718669 DOI: 10.1007/s13577-024-01161-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Acute injury and secondary injury caused by traumatic brain injury (TBI) seriously threaten the health of patients. The purpose of this study was to investigate the role of β-Asarone in TBI-induced neuroinflammation and injury. In this work, the effects of β-Asarone on nerve injury and neuronal apoptosis were investigated in mice with TBI by controlled cortical impingement. The results of this research implied that β-Asarone dose-dependently decreased the mNSS score, brain water content and neuronal apoptosis, but increased the levels of the axonal markers Nrp-1 and Tau in TBI mice. In addition, β-Asarone caused a decrease in the levels of Fas, FasL, and inflammatory factors in cerebrospinal fluid and serum of TBI mice. Therefore, β-Asarone inhibited neuroinflammation and promoted axon regeneration in TBI mice. Besides, β-Asarone treatment inhibited M1 phenotype polarization but promoted M2 phenotype polarization in microglia of TBI mice. Overexpression of Fas and FasL reversed the above effects of β-Asarone. Thus, β-Asarone regulated microglial M1/M2 polarization balance in TBI mice by suppressing Fas/FasL signaling axis. In conclusion, β-Asarone inhibited Fas/FasL signaling pathway to promote the M1/M2 polarization balance of microglia toward M2 polarization, thus alleviating TBI-induced nerve injury.
Collapse
Affiliation(s)
- Mingyue Xia
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Chunyuan Guo
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- Jiangxi Provincial, People's Hospital, Clinical College of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yeli Xie
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
| | - Wenting Yu
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
| | - Dongsheng Wang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingping Dai
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China.
| |
Collapse
|
43
|
Cáceres E, Olivella JC, Di Napoli M, Raihane AS, Divani AA. Immune Response in Traumatic Brain Injury. Curr Neurol Neurosci Rep 2024; 24:593-609. [PMID: 39467990 PMCID: PMC11538248 DOI: 10.1007/s11910-024-01382-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW This review aims to comprehensively examine the immune response following traumatic brain injury (TBI) and how its disruption can impact healing and recovery. RECENT FINDINGS The immune response is now considered a key element in the pathophysiology of TBI, with consequences far beyond the acute phase after injury. A delicate equilibrium is crucial for a healthy recovery. When this equilibrium is disrupted, chronic inflammation and immune imbalance can lead to detrimental effects on survival and disability. Globally, traumatic brain injury (TBI) imposes a substantial burden in terms of both years of life lost and years lived with disability. Although its epidemiology exhibits dynamic trends over time and across regions, TBI disproportionally affects the younger populations, posing psychosocial and financial challenge for communities and families. Following the initial trauma, the primary injury is succeeded by an inflammatory response, primarily orchestrated by the innate immune system. The inflammasome plays a pivotal role during this stage, catalyzing both programmed cell death pathways and the up-regulation of inflammatory cytokines and transcription factors. These events trigger the activation and differentiation of microglia, thereby intensifying the inflammatory response to a systemic level and facilitating the migration of immune cells and edema. This inflammatory response, initially originated in the brain, is monitored by our autonomic nervous system. Through the vagus nerve and adrenergic and cholinergic receptors in various peripheral lymphoid organs and immune cells, bidirectional communication and regulation between the immune and nervous systems is established.
Collapse
Affiliation(s)
- Eder Cáceres
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia.
- School of Medicine, Universidad de La Sabana, Chía, Colombia.
- Bioscience PhD. School of Engineering, Universidad de La Sabana, Chía, Colombia.
| | | | - Mario Di Napoli
- Neurological Service, SS Annunziata Hospital, Sulmona, L'Aquila, Italy
| | - Ahmed S Raihane
- School of Medicine, University of New Mexico, Albuquerque, NM, USA
- Department of Neurology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| |
Collapse
|
44
|
Genrikhs EE, Shedenkova MO, Voronkov DN, Isaev NK, Stelmashook EV. Activation of Microglia and Astroglia in Unilateral Focal Traumatic Brain Injury in Rats. Bull Exp Biol Med 2024; 178:196-201. [PMID: 39762704 DOI: 10.1007/s10517-025-06306-0] [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: 05/22/2024] [Indexed: 01/15/2025]
Abstract
The number of microglia cells and astrocytes in layer V of the cerebral cortex was estimated on day 7 after damage caused by a unilateral focal traumatic brain injury of the left hemisphere sensorimotor cortex. Quantitative assessment was performed by counting immunocytochemically stained microglia cells (Iba1 marker) and activated astrocytes (GFAP) at different distances from the lesion site. Activation of microglial and astroglial cells was observed not only in the marginal zone of the lesion of the left hemisphere, but also in the intact hemisphere. The data obtained indicate the dissemination of inflammation in focal traumatic brain injury.
Collapse
Affiliation(s)
| | | | | | - N K Isaev
- Research Center of Neurology, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | |
Collapse
|
45
|
Gupta A, Bice Z, Chen V, Chen Y, Veltri AJ, Lin CW, Ma X, Pan AY, Zennadi R, Palecek SP, Mohieldin AM, Nauli SM, Ramchandran R, Rarick KR. Severe traumatic brain injury temporally affects cerebral blood flow, endothelial cell phenotype, and cilia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.623875. [PMID: 39605741 PMCID: PMC11601676 DOI: 10.1101/2024.11.19.623875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Previous clinical work suggested that altered cerebral blood flow (CBF) in severe traumatic brain injury (sTBI) correlates with poor executive function and clinical outcome. However, the molecular consequences of altered CBF on endothelial cells (ECs) and their blood flow-sensor organelle called cilia are not known. Methods We performed laser speckle contrast imaging, single cell isolation, and single cell RNA sequencing (scRNAseq) after sTBI in a closed skull, linear impact mouse model. Validation of select ciliary target protein changes was performed using flow cytometry. Additionally, in vitro experiments modeled the post-injury hypoxic environment to evaluate the effect on cilia protein ARL13B in human brain microvascular ECs. Results We detected immediate reductions in CBF that were sustained for at least 100 minutes in both impacted and non-impacted sides of the brain. Our scRNAseq data detected heterogeneity in the brain cortex-derived EC cluster and demonstrated that two of five unique EC sub-clusters changed their relative proportions post-sTBI. Consistent with flow changes, we identified multiple genes associated with the fluid shear stress pathway that were significantly differentially expressed in brain ECs post-injury. Also, ECs displayed activation of ischemic pathway as early as day 1 post-injury, and enrichment of hypoxia pathway at day 7 and 28 post- injury. Arl13b ciliary gene expression was lost on day 1 in ECs cluster and remained lost for the entire course of the injury. We validated the loss of cilia protein ARL13B specifically from brain ECs as early as day 1 post-injury and detected the protein in the peripheral blood of the injured mice. We also determined that hypoxia could induce loss of ARL13B protein from cultured ECs. Conclusions In severe TBI, blood flow is disrupted in both impacted and non-impacted regions of the brain, creating a hypoxic environment that may influence ciliary gene and protein expression on ECs.
Collapse
|
46
|
Calderone A, Latella D, Cardile D, Gangemi A, Corallo F, Rifici C, Quartarone A, Calabrò RS. The Role of Neuroinflammation in Shaping Neuroplasticity and Recovery Outcomes Following Traumatic Brain Injury: A Systematic Review. Int J Mol Sci 2024; 25:11708. [PMID: 39519259 PMCID: PMC11546226 DOI: 10.3390/ijms252111708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroplasticity and neuroinflammation are variables seen during recovery from traumatic brain injury (TBI), while biomarkers are useful in monitoring injury and guiding rehabilitation efforts. This systematic review examines how neuroinflammation affects neuroplasticity and recovery following TBI in animal models and humans. Studies were identified from an online search of the PubMed, Web of Science, and Embase databases without any search time range. This review has been registered on Open OSF (n) UDWQM. Recent studies highlight the critical role of biomarkers like serum amyloid A1 (SAA1) and Toll-like receptor 4 (TLR4) in predicting TBI patients' injury severity and recovery outcomes, offering the potential for personalized treatment and improved neurorehabilitation strategies. Additionally, insights from animal studies reveal how neuroinflammation affects recovery, emphasizing targets such as NOD-like receptor family pyrin domain-containing 3 (NLRP3) and microglia for enhancing therapeutic interventions. This review emphasizes the central role of neuroinflammation in TBI, and its adverse impact on neuroplasticity and recovery, and suggests that targeted anti-inflammatory treatments and biomarker-based personalized approaches hold the key to improvement. Such approaches will need further development in future research by integrating neuromodulation and pharmacological interventions, along with biomarker validation, to optimize management in TBI.
Collapse
Affiliation(s)
- Andrea Calderone
- Department of Clinical and Experimental Medicine, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy
| | - Desirèe Latella
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Davide Cardile
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Antonio Gangemi
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Francesco Corallo
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Carmela Rifici
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Angelo Quartarone
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| |
Collapse
|
47
|
Zhai X, Wang Z, Gao J. Quercetin alleviates microglial-induced inflammation after traumatic brain injury via the PGC-1α/Nrf2 pathway dependent on HDAC3 inhibition. Brain Res Bull 2024; 217:111080. [PMID: 39277018 DOI: 10.1016/j.brainresbull.2024.111080] [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: 07/25/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Inflammation and neuronal apoptosis play a key role in traumatic brain injury (TBI). Quercetin (Que) has been shown to exhibit a neuroprotective effect after TBI, but the underlying molecular mechanism remains unclear. In this study, We established a weight-drop mouse model to illustrate the effects of Que on microglial-induced inflammation in TBI. Mice were divided into four groups: the Sham group, TBI group, TBI+vehicle group, and TBI+Que group. The TBI+Que group was treated with Que 30 min after TBI. Brain water content, neurological score, and neuronal apoptosis were measured. Western blotting, TUNEL staining, Nissl staining, quantitative polymerase chain reaction, and immunofluorescence staining were performed to assess the activation of the PGC-1α/Nrf2 pathway and nuclear translocation of HDAC3 with Que treatment. The results showed that Que administration alleviated TBI-induced neurobehavioral deficits, encephaledema, and neuron apoptosis. Que also restrained TBI-induced microglial activity and the subsequent expression of the inflammatory factor in the contusion cortex. Moreover, Que treatment activated the PGC-1α/Nrf2 pathway, attributable to the inhibition of HDAC3 translocation to the nucleus. Overall, these results reveal the role of Que in protecting against TBI-induced neuroinflammation and promoting neurological functional recovery, which is achieved through the negative regulation of HDAC3.
Collapse
Affiliation(s)
- Xiaofu Zhai
- Department of Neurosurgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China; Department of Neurosurgery, Lianshui People's Hosptial of Kangda College Affiliated to Nanjing Medical University, Huai'an, Jiangsu 223499, China
| | - Ziyu Wang
- Department of Neurosurgery, The Second People's Hospital of Huai'an, Xuzhou Medical College, Huai'an, Jiangsu 223002, China
| | - Juemin Gao
- Department of Neurosurgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China.
| |
Collapse
|
48
|
Zhang Y, Li T, Wang G, Ma Y. Advancements in Single-Cell RNA Sequencing and Spatial Transcriptomics for Central Nervous System Disease. Cell Mol Neurobiol 2024; 44:65. [PMID: 39387975 PMCID: PMC11467076 DOI: 10.1007/s10571-024-01499-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
The incidence of central nervous system (CNS) disease has persistently increased over the last several years. There is an urgent need for effective methods to improve the cure rates of CNS disease. However, the precise molecular basis underlying the development and progression of major CNS diseases remains elusive. A complete molecular map will contribute to research on CNS disease treatment strategies. Emerging technologies such as single-cell RNA sequencing (scRNA-seq) and Spatial Transcriptomics (ST) are potent tools for exploring the molecular complexity, cell heterogeneity, and functional specificity of CNS disease. scRNA-seq and ST can provide insights into the disease at cellular and spatial transcription levels. This review presents a survey of scRNA-seq and ST studies on CNS diseases, such as chronic neurodegenerative diseases, acute CNS injuries, and others. These studies offer novel perspectives in treating and diagnosing CNS diseases by discovering new cell types or subtypes associated with the disease, proposing new pathophysiological mechanisms, uncovering novel therapeutic targets, and identifying putative biomarkers.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pharmacy, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Teng Li
- Department of Laboratory Medicine, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Yabin Ma
- Department of Pharmacy, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
49
|
Tastan B, Heneka MT. The impact of neuroinflammation on neuronal integrity. Immunol Rev 2024; 327:8-32. [PMID: 39470038 DOI: 10.1111/imr.13419] [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] [Indexed: 10/30/2024]
Abstract
Neuroinflammation, characterized by a complex interplay among innate and adaptive immune responses within the central nervous system (CNS), is crucial in responding to infections, injuries, and disease pathologies. However, the dysregulation of the neuroinflammatory response could significantly affect neurons in terms of function and structure, leading to profound health implications. Although tremendous progress has been made in understanding the relationship between neuroinflammatory processes and alterations in neuronal integrity, the specific implications concerning both structure and function have not been extensively covered, with the exception of perspectives on glial activation and neurodegeneration. Thus, this review aims to provide a comprehensive overview of the multifaceted interactions among neurons and key inflammatory players, exploring mechanisms through which inflammation influences neuronal functionality and structural integrity in the CNS. Further, it will discuss how these inflammatory mechanisms lead to impairment in neuronal functions and architecture and highlight the consequences caused by dysregulated neuronal functions, such as cognitive dysfunction and mood disorders. By integrating insights from recent research findings, this review will enhance our understanding of the neuroinflammatory landscape and set the stage for future interventions that could transform current approaches to preserve neuronal integrity and function in CNS-related inflammatory conditions.
Collapse
Affiliation(s)
- Bora Tastan
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| |
Collapse
|
50
|
Ke JP, He BD, Gong ML, Yan ZZ, Du HZ, Teng ZQ, Liu CM. Loss of microglial Arid1a exacerbates microglial scar formation via elevated CCL5 after traumatic brain injury. Cell Commun Signal 2024; 22:467. [PMID: 39350161 PMCID: PMC11443815 DOI: 10.1186/s12964-024-01852-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Traumatic brain injury (TBI) is an acquired insult to the brain caused by an external mechanical force, potentially resulting in temporary or permanent impairment. Microglia, the resident immune cells of the central nervous system, are activated in response to TBI, participating in tissue repair process. However, the underlying epigenetic mechanisms in microglia during TBI remain poorly understood. ARID1A (AT-Rich Interaction Domain 1 A), a pivotal subunit of the multi-protein SWI/SNF chromatin remodeling complex, has received little attention in microglia, especially in the context of brain injury. In this study, we generated a Arid1a cKO mouse line to investigate the potential roles of ARID1A in microglia in response to TBI. We found that glial scar formation was exacerbated due to increased microglial migration and a heightened inflammatory response in Arid1a cKO mice following TBI. Mechanistically, loss of ARID1A led to an up-regulation of the chemokine CCL5 in microglia upon the injury, while the CCL5-neutralizing antibody reduced migration and inflammatory response of LPS-stimulated Arid1a cKO microglia. Importantly, administration of auraptene (AUR), an inhibitor of CCL5, repressed the microglial migration and inflammatory response, as well as the glial scar formation after TBI. These findings suggest that ARID1A is critical for microglial response to injury and that AUR has a therapeutic potential for the treatment of TBI.
Collapse
Affiliation(s)
- Jin-Peng Ke
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Bao-Dong He
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Mao-Lei Gong
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zhong-Ze Yan
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hong-Zhen Du
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zhao-Qian Teng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Chang-Mei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| |
Collapse
|