1
|
Song Y, Wang L, Peng T, Shan L, Wan B, Tang M, Luan Y, Jiang Y, He W. Brain-targeting biomimetic nanozyme enhances neuroprotection in ischemic stroke by remodeling the neurovascular unit. J Control Release 2025; 382:113750. [PMID: 40254137 DOI: 10.1016/j.jconrel.2025.113750] [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: 01/30/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Dysfunction of the neurovascular unit significantly impacts the prognostic outcomes of ischemic stroke. However, effective strategies to comprehensively modulate the neurovascular unit have yet to be developed. In this work, we introduce a brain-targeting biomimetic nanozyme, A@HPB@THSA, designed to mitigate neurovascular unit dysfunction following ischemia/reperfusion. Specifically, aspirin is encapsulated within a hollow Prussian blue nanozyme, which is subsequently modified with brain-targeting T7 peptide-conjugated human serum albumin, ultimately forming the composite A@HPB@THSA. The overexpression of transferrin receptors on cerebral vascular endothelial cells, along with compromised blood-brain barrier (BBB) permeability, facilitates the accumulation of A@HPB@THSA at cerebral ischemic lesions. The hollow Prussian blue nanozyme component effectively scavenges reactive oxygen species in ischemia/reperfusion-affected brain cells, while the aspirin component inhibits platelet aggregation and neutrophil infiltration, thereby preventing microvascular "no-reflow" and preserving the integrity of the BBB. In rat models of transient middle cerebral artery occlusion, A@HPB@THSA demonstrated comprehensive modulation of the neurovascular unit, including reduced BBB permeability, promoted microglia polarization toward an anti-inflammatory phenotype, and enhanced neuronal survival. This work provides a promising platform to reverse dysfunctional neurovascular unit for ischemic stroke treatment.
Collapse
Affiliation(s)
- Yan Song
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Luyao Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tingting Peng
- Departent of Pharmacy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Lingling Shan
- Departent of Pharmacy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Bo Wan
- Departent of Pharmacy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Mingtan Tang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuxia Luan
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yue Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Wenxiu He
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
2
|
Liu Y, Takamatsu Y, Chen K, Ding Y, Oka Y, Sugiyama T, Maejima H. Skilled reaching training combined with pharmacological inhibition of histone deacetylases potentiated motor recovery after intracerebral hemorrhage in a synergic manner. Brain Res 2025; 1856:149569. [PMID: 40081517 DOI: 10.1016/j.brainres.2025.149569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Neuronal recovery after stroke is supported by the expression of genes involved in neuronal plasticity and neuroprotection. As an epigenetic modification, histone acetylation modulates gene expression elicited by neurorehabilitation. This study aimed to investigate the combined effects of skilled reaching training (SRT) and the pharmacological inhibition of histone deacetylase (HDAC) using sodium butyrate (NaB) on motor function recovery after intracerebral hemorrhage (ICH). Wistar rats were divided into five groups: Sham, ICH, ICH plus SRT (ICH + SRT), ICH plus NaB administration (ICH + NaB), and ICH plus SRT plus NaB administration (ICH + SRT + NaB). ICH surgery was conducted based on the microinjection of collagenase into the striatum near the internal capsule. NaB treatment (300 mg/kg injected intraperitoneally) and SRT were performed five days a week for four weeks after ICH surgery, followed by tissue collection. After the intervention, the ICH + SRT + NaB group exhibited significant improvement in skilled motor function, accompanied by a significant increase in neurotrophin 4 and synaptophysin expression in the ipsilateral motor cortex. This study showed that combination therapy of SRT and HDAC inhibition synergistically promoted motor recovery after ICH, accompanied by the upregulation of crucial genes for neuroplasticity. Taken together, this study indicates that HDAC inhibition could represent an enriched neuronal platform for neurorehabilitation after ICH.
Collapse
Affiliation(s)
- Yushan Liu
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Yasuyuki Takamatsu
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan; Department of Physical Therapy, College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai 487-8501, Japan
| | - Ke Chen
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Yuan Ding
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Yuichiro Oka
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Takuya Sugiyama
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroshi Maejima
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
3
|
Yamamoto S, Shiraishi K, Kushida Y, Oguma Y, Wakao S, Dezawa M, Kuroda S. Nose-to-brain delivery of human muse cells enhances structural and functional recovery in the murine ischemic stroke model. Sci Rep 2025; 15:16243. [PMID: 40346279 PMCID: PMC12064699 DOI: 10.1038/s41598-025-96451-3] [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: 01/27/2025] [Accepted: 03/28/2025] [Indexed: 05/11/2025] Open
Abstract
Muse cells are endogenous, non-tumorigenic, pluripotent-like stem cells already applied to clinical trials based on intravenous injection. They can selectively home to the post-infarct area, replenish apoptotic neural cells by phagocytosis-induced differentiation, and enhance functional recovery. The effect of nose-to-brain delivery of Muse cells on cerebral infarct was examined. Permanent middle cerebral artery occlusion model BALB/c mice received intranasal administration of either human Muse cells (6.0 × 104 cells), high-dose human-mesenchymal stem cells (MSCs) (1.6 × 106 cells), low-dose human-MSCs (6.0 × 104 cells), or vehicle at 7 days after onset. An accelerated rotarod test and a histological assessment were done. The vehicle- or low-dose MSC groups showed no significant improvement in the rotarod test. In the high-dose MSC group, motor function was transiently recovered, but the therapeutic effect disappeared thereafter. The Muse group continuously improved motor function, with statistical significance to the other groups. The engraftment of administered cells in the peri-infarct area was the highest in the Muse group, while few cells were detected in other groups. 63.6 ± 8.5% and 26.2 ± 3.0% of Muse cells were positive for NeuN and GSTpi, respectively. Intranasal administration of Muse cells might be a viable approach to improving functional recovery with less invasiveness after ischemic stroke.
Collapse
Affiliation(s)
- Shusuke Yamamoto
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Keitaro Shiraishi
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yo Oguma
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Satoshi Kuroda
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
4
|
Han J, Xu Y, Zhou Y, Zhu N, Gao J, Huang L. Therapeutic Hypothermia and Recombinant Erythropoietin Mitigate Brain Microvascular Endothelial Cell Dysfunction via Modulating the Pentose Phosphate Pathway. J Mol Neurosci 2025; 75:65. [PMID: 40343581 DOI: 10.1007/s12031-025-02356-1] [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: 03/20/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Neonatal brains are particularly vulnerable to oxidative stress, making the pentose phosphate pathway (PPP) pivotal in damage limitation. This study aimed to confirm the mechanism of erythropoietin combined with therapeutic hypothermia (TH) in hypoxic-ischemic brain damage (HIBD). Neonatal HIBD rat models were employed and the impacts of erythropoietin and TH on behavior, cerebral infarction, pathology, and microvascular were evaluated. Following that, the assessments of inflammation, oxidative stress, apoptosis, and the level of glucose-6-phosphate dehydrogenase (G6PD, rate-limiting enzyme in the PPP) proceeded. Human brain microvascular endothelial cells (HBMECs) underwent oxygen-glucose deprivation (OGD) and were treated with TH and G6PD inhibitor RRx-001. The impacts of the G6PD inhibitor on HBMEC function and barrier were evaluated. Simultaneous administration of TH and EPO reduced pathological damage and attenuated microvascular loss. In addition, this combination therapy had anti-inflammatory, antioxidant, and anti-apoptotic properties, and enhanced G6PD activity, both in vivo and in vitro. Inhibition of G6PD disrupted the protective effects of TH and EPO on the patency of the PPP and the function of HBMECs, and barrier integrity was further broken. This study reveals that the combination of TH and EPO mitigates microvascular endothelial cell dysfunction via partially modulating the PPP, thus preserving barrier integrity.
Collapse
Affiliation(s)
- Jinan Han
- Department of Pediatrics, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Ying Xu
- Department of Pediatrics, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Yan Zhou
- Department of Pediatrics, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Ning Zhu
- Department of Pediatrics, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Jiazhen Gao
- Department of Pediatrics, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Li Huang
- Department of Pediatrics, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
5
|
da Silva Joaquim L, da Rosa LR, Strickert Y, Machado RS, Lanzzarin E, Bernardes G, de Souza Ramos S, de Novais LR, Steiner B, Farias B, Mathias K, Martins HM, Lins EMF, Chaves JS, Camilo D, da Silva LE, de Oliveira MP, da Silva MR, Barcelos PMP, Santos FP, Bobinski F, Rezin GT, Yonamine M, Inserra A, Petronilho F, de Bitencourt RM. Ayahuasca reverses ischemic stroke-induced neuroinflammation and oxidative stress. Behav Brain Res 2025; 485:115521. [PMID: 40043852 DOI: 10.1016/j.bbr.2025.115521] [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: 09/25/2024] [Revised: 02/21/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Ischemic stroke is a leading cause of death and disability worldwide. Survivors face disability and psychiatric sequelae resulting from ischemia-induced cell death and associated neuroinflammation, and oxidative stress. Herbal medicines have been shown to elicit neuroprotective effects following stroke due to their anti-inflammatory and antioxidant effects. Preliminary evidence suggests that Ayahuasca (AYA), a decoction made from the vine Banisteriopsis caapi containing β-carbolines and the shrub Psychotria viridis containing N, N-Dimethyltryptamine, might attenuate ischemia-induced neuroinflammation and oxidative stress. Therefore, in this study we investigated the putative protective effects of AYA in the middle cerebral artery occlusion (MCAO) model of ischemic stroke. METHODS Wistar rats were subjected to the MCAO stroke model or sham surgery on day 0. After 24-h, rats were treated for three days with AYA (2 and 4 mL/kg, gavage) or saline. Neurological score was assessed for 72-h post-stroke. Rats were tested in the elevated plus maze, open field, and novel object recognition tests to assess locomotion, anxiety-like behavior, and recognition memory. Interleukin (IL)-6, IL-10 myeloperoxidase (MPO) activity, and the nitrite/nitrate (N/N) concentrations were determined in the prefrontal cortex (PFC), hippocampus (HPC), hypothalamus (HYP) and cortex. as markers of inflammation. Oxidative stress was quantified in the same brain areas as measured by the levels of thiobarbituric acid reactive species (TBARS), protein carbonylation, and superoxide dismutase (SOD), and catalase (CAT) activity. Mitochondrial metabolism was assessed quantifying the activity of complex 1(CI), CII, citrate synthase (CS), succinate dehydrogenase (SDH), and creatine kinase (CK). RESULTS No differences were observed regarding neurological deficits, locomotion, anxiety-like behavior, and recognition memory. However, AYA reversed the stroke-induced increase in IL-6 levels in the PFC and the HPC, IL-10 in the PFC, HPC, and HYP, MPO activity in the PFC, and N/N concentration and CAT activity in the HYP. Moreover, AYA decreased TBARS levels in the PFC and HPC and brain-derived neurotrophic factor (BDNF) in the PFC, and increased SOD activity in the cortex. Lastly, AYA increased CI activity in the HPC and cortex and decreased SDH and CK activity in the HPC. CONCLUSION AYA administration following ischemic stroke modulates oxidative stress and neuroinflammation in the PFC, HPC, and HYP. Despite no significant improvements in neurological or behavioral scores, these molecular changes suggest a neuroprotective role of AYA. Future studies should explore the timing of AYA treatment and putative long-term effects on functional recovery, as well as its potential in other brain regions critical for cognitive and motor functions.
Collapse
Affiliation(s)
- Larissa da Silva Joaquim
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Lara Rodrigues da Rosa
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Yasmin Strickert
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil; Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Everton Lanzzarin
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Gabriela Bernardes
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Suelen de Souza Ramos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Linério Ribeiro de Novais
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Beatriz Steiner
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Brenno Farias
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Helena Mafra Martins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Elisa Mitkus Flores Lins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Jéssica Schaefer Chaves
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Douglas Camilo
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Pablo Michel Pereira Barcelos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabiana Pereira Santos
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Maurício Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio Inserra
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Rafael Mariano de Bitencourt
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil.
| |
Collapse
|
6
|
Wang W, Li K, Ma W, Li Y, Liu F, Kong Y, Wang L, Yi F, Sang Y, Li G, Liu H, Qiu J. Ultrasound-activated piezoelectric nanostickers for neural stem cell therapy of traumatic brain injury. NATURE MATERIALS 2025:10.1038/s41563-025-02214-w. [PMID: 40329084 DOI: 10.1038/s41563-025-02214-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 03/17/2025] [Indexed: 05/08/2025]
Abstract
Traumatic brain injury (TBI) is associated with life-threatening and permanent disabilities. Given the limited capacity of neurons to regenerate, effective treatments for TBI are lacking. Neural stem cells (NSCs) can differentiate into fully functioning neurons and thus hold promise for TBI treatment. Nonetheless, NSC differentiation and proliferation are slow and inefficient. Studies have shown that piezoelectric stimulation is capable of promoting the differentiation and proliferation of NSCs. Here, we describe barium titanate-reduced graphene oxide (BTO/rGO) hybrid piezoelectric nanostickers that promote NSC proliferation and differentiation. These hybrid nanostickers attach to NSC membranes, serving as long-term generators of piezoelectric potentials upon ultrasound stimulation. BTO/rGO nanostickers promote rapid neuronal differentiation and maturation by activating the voltage-gated calcium channel/Ca2+/calmodulin-dependent protein kinase II/cAMP response element-binding protein pathways. Transplantation of NSCs with BTO/rGO nanostickers into the injured brain region of rats with TBI substantially repairs brain tissue and effectively restores physiological functions after 28 d following 5-min ultrasound irradiation every 2 d. These results demonstrate the potential of the combination of NSCs and BTO/rGO nanostickers for TBI treatment.
Collapse
Affiliation(s)
- Wenhan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Keyi Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Wenjun Ma
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Yiwei Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Feng Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Ying Kong
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Liang Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China.
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China.
- Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, China.
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China.
| |
Collapse
|
7
|
Yang L, Gao Y, Lopes LS, Lian J, Fu W, Tan H, Yang S, Xie Z, Huang Y, Zhang J, Lu Y, Tang H, Xiong B, Wei X, Xie L, Peng Y, Liu X, Han H. Accelerated Molecular Transportation in the Brain Extracellular Space with 755-nm Light Attenuates Post-Stroke Cognitive Impairment in Rats. CYBORG AND BIONIC SYSTEMS 2025; 6:0262. [PMID: 40330544 PMCID: PMC12053100 DOI: 10.34133/cbsystems.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 05/08/2025] Open
Abstract
Ischemic stroke exacts a heavy toll in death and disability worldwide. After ischemic stroke, the accumulation of pathobiomolecules in the brain extracellular space (ECS) will exacerbate neurological damage and cognitive impairment. Photobiomodulation (PBM) has been demonstrated to improve cognitive function in Alzheimer's disease mouse models by accelerating molecular transportation in the brain ECS. This suggests that PBM may have a potential role in the accumulation of pathobiomolecules in the brain ECS following ischemic stroke. In this study, we developed a PBM therapy apparatus with custom parameters. By evaluating the treatment effect, we identified that 755 nm was the optimal light wavelength for ischemic stroke in rats with transient middle cerebral artery occlusion/reperfusion. Extracellular diffusion and interstitial fluid (ISF) drainage were measured using a tracer-based magnetic resonance imaging method. Our results showed that PBM accelerated molecular transportation in the brain ECS and ISF drainage, promoting the clearance of pro-inflammatory cytokines and reducing the deposition of pathological proteins. Consequently, the infarct volume decreased and neurological cognitive function was improved. Besides, the acceleration of ISF drainage was achieved by reducing expression and restoring polarization of aquaporin 4 (AQP4) in the peri-infarct area. In summary, we demonstrated that PBM could alleviate ischemia-reperfusion injury and prevent post-stroke cognitive impairment by accelerating molecular transportation in the brain ECS, paving a pathway for ischemic stroke treatment via the light-ECS interaction.
Collapse
Affiliation(s)
- Liu Yang
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Yajuan Gao
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Leonor Serrano Lopes
- Department of Nuclear Medicine, Inselspital, Bern University Hospital,
University of Bern, Bern 3010, Switzerland
- Graduate School for Cellular and Biomedical Sciences,
University of Bern, Bern 3012, Switzerland
| | - Jingge Lian
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Wanyi Fu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
- Department of Electronic Engineering,
Tsinghua University, Beijing 100084, China
| | - Hanbo Tan
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Shuangfeng Yang
- Department of Radiology, Beijing Children’s Hospital,
Capital Medical University, National Center for Children’ Health, Beijing 100045, China
| | - Zhaoheng Xie
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Yixing Huang
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Jicong Zhang
- School of Biological Science and Medical Engineering,
Beihang University, Beijing 100191, China
| | - Yanye Lu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Hao Tang
- School of Computer Science,
Peking University, Beijing 100871, China
| | - Bo Xiong
- School of Computer Science,
Peking University, Beijing 100871, China
| | - Xunbin Wei
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Lide Xie
- Chengde Medical University, Chengde, Hebei 067000, China
| | - Yun Peng
- Department of Radiology, Beijing Children’s Hospital,
Capital Medical University, National Center for Children’ Health, Beijing 100045, China
| | - Xinyu Liu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Hongbin Han
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
- Chengde Medical University, Chengde, Hebei 067000, China
| |
Collapse
|
8
|
Feng Q, Li Y, Wen X, Li H, Qi C, Wang N, Zhu G, Fu Y, Liu C, Liu D, Zhang Z, Yang F, Zhou Z, Song J, Liang J, Chen Y, Zhou X, Liu Y, Song Y. TAT-T407 Mitigates Apoptosis and Cognitive Impairments Following Cerebral Ischemia Through Disruption of TRPV1-CDK5 Interaction. Mol Neurobiol 2025:10.1007/s12035-025-04926-1. [PMID: 40317415 DOI: 10.1007/s12035-025-04926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/05/2025] [Indexed: 05/07/2025]
Abstract
Cerebral ischemia/reperfusion (I/R) injury manifests as progressive motor and cognitive dysfunction, primarily attributed to neuronal apoptosis. However, there is a lack of neuroprotective drugs targeting neuronal apoptosis in ischemic stroke. In this study, utilizing bioinformatics analysis, we hypothesized that TRPV1 could serve as a novel molecular target implicated in neuronal apoptosis during cerebral ischemia/reperfusion (I/R) injury. To validate our hypothesis in vivo, we employed mouse models of I/R injury induced by transient middle cerebral artery occlusion (tMCAO). Importantly, pre-injecting capsazepine (CPZ), a TRPV1 antagonist, significantly suppressed apoptotic pathway activity in neurons. Additionally, we investigated the regulatory role of CDK5, a well-known neuronal-specific kinase, in modulating the internalization and functionality of TRPV1 ion channels. Our findings revealed an augmented interaction between TRPV1 and CDK5 during cerebral ischemia/reperfusion (I/R) injury. The administration of the TAT-T407 interference peptide, derived from the phosphorylation site of TRPV1 for CDK5, resulted in a reduction of neuronal apoptosis within ischemic regions following cerebral ischemia/reperfusion (I/R) injury. This intervention significantly diminished cerebral infarct volume and improved neurological function. In summary, disrupting the TRPV1/CDK5 interaction through TAT-T407 peptides provides protection against neuronal apoptosis and cognitive decline, suggesting an innovative therapeutic strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Qian Feng
- Jiangsu Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Clinical Laboratory, Xuzhou Medical University Affiliated Hospital, Xuzhou, Jiangsu, 221002, China
| | - Ying Li
- Clinical Laboratory, Xuzhou Medical University Affiliated Hospital, Xuzhou, Jiangsu, 221002, China
| | - Xiangru Wen
- Jiangsu Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Hui Li
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, 999077, China
| | - Chengyu Qi
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Nan Wang
- Jiangsu Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, 999077, China
| | - Yanyan Fu
- Department of Neurobiology and Cell Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Changdong Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, 999077, China
| | - Dan Liu
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Zhen Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Fan Yang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Zhongyuan Zhou
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Jinjin Song
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Jia Liang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yuling Chen
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Xiaoyan Zhou
- Jiangsu Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Yan Liu
- Jiangsu Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Yuanjian Song
- Jiangsu Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Engineering Center for Precision Diagnosis and Treatment Research of Polygenic Diseases, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
9
|
Pan S, Wen Y, Liu Z, Xu K, Zhang N, Tong X, Teng Y, Song X, Tong X. Neuroprotective effects of bone marrow mesenchymal stem cells combined with mannitol on radiation-induced brain injury by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. Brain Res Bull 2025; 224:111326. [PMID: 40174787 DOI: 10.1016/j.brainresbull.2025.111326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/07/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) have demonstrated potential in the treatment of radiation-induced brain injury (RIBI); however, the presence of the blood-brain barrier (BBB) limits their therapeutic efficacy. Additionally, the precise mechanisms behind the use of BMSCs in treating RIBI are still not well understood. This study aimed to investigate the therapeutic efficacy of mannitol and BMSCs on neuronal autophagy and their efficacy in treating RIBI. METHODS In the study, RIBI models were first established in male Sprague-Dawley (SD) rats. Evans blue staining was performed to examine how mannitol influences BBB permeability in RIBI. We isolated BMSCs from SD rats using the whole bone marrow adherent method and assessed their adipogenic and osteogenic differentiation potential through Oil Red O and Alizarin Red S staining; flow cytometry analysis assessed cell surface markers. Prussian blue staining was employed to verify the migration of iron-labeled BMSCs into brain tissue. The rats were then divided into specific treatment groups, and model establishment followed according to experimental conditions. The body weight of the rats was measured weekly throughout the study. Cognitive function was assessed using the Morris water maze test. H&E and Nissl staining were applied to evaluate hippocampal neuronal survival. We quantified key proteins in the PI3K/AKT/mTOR signaling pathway by Western blotting, and quantified autophagy-related proteins LC3B and beclin-1 using both Western blotting and immunofluorescence. RESULTS Mannitol treatment significantly increased BBB permeability and promoted BMSCs migration. The combination of BMSCs and mannitol improved cognitive and memory functions, leading to better body weight recovery compared to the BMSCs group. H&E and Nissl staining also revealed a significant increase in neuronal survival within the combined treatment group. Furthermore, we observed through Western blot and immunofluorescence analyses that combination of BMSCs and mannitol enhanced the activation of PI3K, AKT, and mTOR through phosphorylation, while it reduced the expression levels of LC3B and beclin-1. CONCLUSION The combination of BMSCs and mannitol treatment significantly improved cognitive function and hippocampal neuronal survival in RIBI rats. This effect was achieved by increasing BBB permeability, facilitating BMSCs migration to the injured region, and regulating excessive autophagy through the PI3K/AKT/mTOR pathway. This combined treatment demonstrated a neuroprotective effect superior to that of BMSCs treatment alone.
Collapse
Affiliation(s)
- Shichao Pan
- College of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuxin Wen
- College of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Zhanhong Liu
- Department of CT/MRI, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong 637000, China
| | - Kaina Xu
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Na Zhang
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Xingbo Tong
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Ye Teng
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Xue Song
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Xu Tong
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China.
| |
Collapse
|
10
|
Iwabuchi N, Uchida H, Abe T, Kajitani T, Aburakawa D, Mansour A, Endo H, Tominaga T, Niizuma K. Multilineage-differentiating stress-enduring cells attenuate the cognitive impairment caused by chronic cerebral hypoperfusion in rats. Exp Neurol 2025; 387:115185. [PMID: 39952380 DOI: 10.1016/j.expneurol.2025.115185] [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: 09/28/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are non-tumorigenic pluripotent- like stem cells that can migrate to damaged sites and contribute to tissue repair. Chronic cerebral hypoperfusion (CCH), which mimics vascular dementia, causes hippocampal neuronal degeneration and white matter (WM) damage, which lead to cognitive dysfunction. Currently, there are no effective treatments for it. We evaluated the efficiency of the human-Muse cell-based product CL2020 in treating CCH in rats. A bilateral common carotid artery occlusion was used to induce cognitive dysfunction. Six-weeks after carotid artery occlusion, CL2020 were injected intravenously. Cognitive function was assessed using a Barnes circular maze (BCM) at 3 weeks after CL2020 administration. Histological findings and western blots were assessed at 4 weeks after CL2020 administration. BCM assessment indicated recovery in cognitive function in the CL2020-treated group. Compared with the vehicle, CL2020 targeted the hippocampus, where it decreased neuronal loss and WM damage. CL2020 also promoted angiogenesis and suppressed apoptotic cell death. Western blotting of hippocampal samples revealed the downregulation of pro- apoptotic and the upregulation of anti-apoptotic proteins in the CL2020-treated group. In conclusion, intravenous administration of CL2020 improved the cognitive deficits caused by CCH, partly because of decreased hippocampal neuronal loss and WM damage, and increased angiogenesis in the hippocampus.
Collapse
Affiliation(s)
- Naoya Iwabuchi
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroki Uchida
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takatsugu Abe
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Kajitani
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daiki Aburakawa
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ahmed Mansour
- Department of Neurosurgery, Menoufia University Graduate School of Medicine, Menoufia, Egypt
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan; Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
11
|
Li L, Hu H, Jiang W, Mao S, Yang Z, Lan T, Hu X, Fang Y, Xu L, Xu J, Yang Y, Jiang W, Chu L. Artemisinin alleviates ischemic stroke injury and promotes neurogenesis through PPARγ-mediated M2 polarization of microglia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156769. [PMID: 40286750 DOI: 10.1016/j.phymed.2025.156769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/15/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ischemic stroke (IS) remains a challenge in clinical treatment due to limited therapeutic options. While artemisinin (ART), an antimalarial drug, shields against acute IS via anti-inflammatory, antioxidant, and anti-apoptotic properties, the long-term benefits and specific underlying mechanisms have not been fully elucidated. Here, we investigate whether ART ameliorates IS injury and promotes neurogenesis by activating the peroxisome proliferator-activated receptor γ (PPARγ)-dependent M2 microglial polarization. METHODS The experimental models included transient middle cerebral artery occlusion/reperfusion (MCAO/R) in rats and oxygen-glucose deprivation/reoxygenation (OGD/R) in primary microglial cultures to simulate IS. The therapeutic effects of ART were evaluated by neurological functions and infarct volume. PPARγ inhibitor T0070907 (T007) was intraperitoneally injected 24 h following MCAO/R at a dose of 2 mg/kg in vivo and a concentration of 10 μM for 30 min before OGD in vitro. We utilized real-time quantitative polymerase chain reaction (RT-qPCR) along with Western blot analyses to detect the microglia markers and PPARγ. The proliferation and differentiation of neural stem cells (NSCs) both in vivo and in vitro were assessed via immunofluorescence labeling. The neurogenic potential of ART-treated microglia was investigated by conditioned medium. The levels of brain-derived growth factor (BDNF) and insulin-like growth factor-1 (IGF-1) in microglia were measured by immunofluorescence staining and enzyme-linked immunosorbent assay (ELISA). RESULTS ART treatment significantly alleviated short- and long-term neurological deficits and reduced cerebral infarct volume in rats with IS. Experiments conducted both in vivo and in vitro experiments illustrated that ART directed microglia away from the pro-inflammatory M1 state towards the anti-inflammatory M2 state, enhanced neurogenesis, and upregulated the expression of PPARγ, BDNF, and IGF-1. In addition, the conditioned medium from ART-exposed microglia stimulated the proliferation and neuronal differentiation of primary NSCs. However, these positive effects were effectively counteracted by the use of PPARγ inhibitor T0070907 (T007). CONCLUSION Our findings demonstrate that ART ameliorates IS injury and promotes neurogenesis mainly through PPARγ-mediated microglia M2 polarization. Therefore, ART can be considered a potential therapeutic drug for IS.
Collapse
Affiliation(s)
- Lin Li
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Huiqin Hu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Weifeng Jiang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Shihui Mao
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zheng Yang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Ting Lan
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Xiaowei Hu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Yan Fang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Lanxi Xu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jiadong Xu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Yan Yang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Weiru Jiang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Lisheng Chu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
12
|
Li K, Liu Y, Gong J, Li J, Zhao M, Hong C, Zhang Y, He M, Zhu Z, Chen Z, Wang Z. Reactive astrocyte-derived exosomes enhance intracranial lymphatic drainage in mice after intracranial hemorrhage. Fluids Barriers CNS 2025; 22:37. [PMID: 40229887 PMCID: PMC11995599 DOI: 10.1186/s12987-025-00651-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND After intracranial hemorrhage (ICH), the formation of primary hematoma foci leads to the development of secondary brain injury factors such as perihematomal edema (PHE) and accumulation of toxic metabolites, which severely affect the survival and prognosis of patients. The intracerebral lymphatic system, proposed by Jeffrey J. Iliff et al., plays an important role in central nervous system (CNS) fluid homeostasis and waste removal, while reactive astrocyte-derived exosomes have shown therapeutic potential in CNS disorders. Our study focuses on the effects of hemin-treated reactive astrocyte-derived exosomes on the functional integrity of the glymphatic system (GLS) after ICH and their potential mechanism of action in repairing brain injury. METHODS Hemin, an iron-rich porphyrin compound, was used to construct the in vitro model of ICH. Primary astrocytes were treated with complete medium supplemented with different concentrations of hemin to obtain exosomes secreted by them, and mice with ICH induced by the collagenase method were intervened by intranasal administration. Solute clearance efficiency was assessed by intracranial injection of cerebrospinal fluid tracers and fluorescent magnetic beads. Immunofluorescence analysis of Aquaporin 4 (AQP4) polarization and astrocyte proliferation. Magnetic Resonance Imaging was used to visualize and quantify the volume of hematoma foci and PHE, and Western Blot was used to analyze the accumulation of toxic metabolites, while neuronal apoptosis was detected by a combination of TUNEL assay apoptosis detection kit and Nissl staining, and their functional status was analyzed. Gait analysis software was used to detect functional recovery of the affected limb in mice. RESULTS Exosomes from hemin treated astrocytes facilitated the recovery of AQP4 polarization and attenuated astrocyte proliferation around hematoma foci in mice with ICH, thereby promoting the recovery of the GLS. Meanwhile, exosomes from hemin treated astrocytes reduced PHE and toxic protein accumulation, decreased apoptosis of cortical neurons on the affected side, and facilitated recovery of motor function of the affected limb, and these effects were blocked by TGN020, an AQP4-specific inhibitor. CONCLUSIONS Exosomes from hemin treated astrocytes attenuated secondary brain injury and neurological deficits in mice with ICH by promoting the repair of GLS injury.
Collapse
Affiliation(s)
- Kexin Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Jing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Chengyou Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Yuchi Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Mengyao He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Zhenye Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, Tianjin, China
| | - Zhijuan Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
13
|
Zhao M, Wang J, Zhu S, Zhang S, Han C, Tan C, Huang Y, Sun Z, Wang L, Liu J. Human neural stem cell-derived exosomes activate PINK1/Parkin pathway to protect against oxidative stress-induced neuronal injury in ischemic stroke. J Transl Med 2025; 23:402. [PMID: 40188077 PMCID: PMC11971779 DOI: 10.1186/s12967-025-06283-y] [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: 11/25/2024] [Accepted: 02/20/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Mitochondria play a critical role in oxidative stress (OS)-induced neuronal injury during ischemic stroke (IS), making them promising therapeutic targets. Mounting evidence underscores the extraordinary therapeutic promise of exosomes derived from human neural stem cells (hNSCs) in the management of central nervous system (CNS) diseases. Nonetheless, the precise mechanisms by which these exosomes target mitochondria to ameliorate the effects of IS remain only partially elucidated. This study investigates the protective effects of hNSC derived exosomes (hNSC-Exos) on neuronal damage. METHODS Using a rat model of middle cerebral artery occlusion (MCAO) in vivo and OS-induced HT22 cells in vitro. Firstly, our research group independently isolated human neural stem cells (hNSCs) and subsequently prepared hNSC-Exos. In vivo, MCAO rats were restored to blood flow perfusion to simulate ischemia-reperfusion injury, and hNSC-Exos were injected through stereotaxic injection into the brain. Subsequently, the protective effects of hNSC-Exos on MCAO rats were evaluated, including histological studies, behavioral assessments. In vivo, H2O2 was used in HT22 cells to simulate the OS environment in MCAO, and then its protective effects on HT22 were evaluated by co-culturing with hNSC-Exos, including immunofluorescence staining, western blotting (WB), quantitative real time PCR (qRT-PCR). In the process of exploring specific mechanisms, we utilized RNA sequencing (RNA-seq) to detect the potential induction of mitophagy in OS-induced HT22 cells. Afterwards, we employed a series of mitochondrial function assessments and autophagy related detection techniques, including measuring mitochondrial membrane potential, reactive oxygen species (ROS) levels, transmission electron microscopy (TEM) imaging, monodansylcadaverine (MDC) staining, and mCherry-GFP-LC3B staining. In addition, we further investigated the regulatory pathway of hNSC-Exos by using autophagy inhibitor mdivi-1 and knocking out PTEN induced kinase 1 (PINK1) in HT22 cells. RESULTS Administration of hNSC-Exos significantly ameliorated brain tissue damage and enhanced behavioral outcomes in MCAO rats. This treatment led to a reduction in brain tissue apoptosis and facilitated the normalization of impaired neurogenesis and neuroplasticity. Notably, the application of hNSC-Exos in vitro resulted in an upregulation of mitophagy in HT22 cells, thereby remedying mitochondrial dysfunction. We demonstrate that hNSC-Exos activate mitophagy via the PINK1/Parkin pathway, improving mitochondrial function and reducing neuronal apoptosis. CONCLUSIONS These findings suggest that hNSC-Exos alleviate OS-induced neuronal damage by regulating the PINK1/Parkin pathway. These reveals a novel role of stem cell-derived mitochondrial therapy in promoting neuroprotection and suggest their potential as a therapeutic approach for OS-associated CNS diseases, including IS.
Collapse
Affiliation(s)
- Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Shuaiyu Zhu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Shensen Zhang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Chao Han
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Chengcheng Tan
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Yubing Huang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Zhaokai Sun
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China.
| |
Collapse
|
14
|
Wang Y, Wei R, Du L. Butylphthalide protects cerebral infarction in a rat model by regulating netrin-1/deleted in colorectal cancer/vascular endothelial growth factor axis. Neuroreport 2025; 36:327-335. [PMID: 40177830 DOI: 10.1097/wnr.0000000000002153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Acute cerebral infarction (CI) is characterized by acute onset, high disability rate, and high morbidity rate, which seriously threatens the health and safety of people and places a heavy burden on individuals and the country. This study aimed to explore the effects of butylphthalide on nerve cell ferroptosis in CI rats and its underlying mechanisms. Middle cerebral artery occlusion (MCAO) rat model was used to study the effect of butylphthalide on acute CI in vivo and oxygen glucose deprivation (OGD) model was used to study the effect of butylphthalide on acute CI in vitro. Our findings demonstrated that butylphthalide markedly reduced oxidative damage and ferroptosis in the brains of MCAO rats. Furthermore, we found that butylphthalide upregulated the netrin-1/deleted in colorectal cancer (DCC)/vascular endothelial growth factor (VEGF) signaling axis, which regulates NF-E2-related factor-2 (NRF2) expression and contributes to ferroptosis in the MCAO rat model and OGD-treated HT22 cells. Collectively, our findings indicate that butylphthalide inhibits oxidative stress-mediated ferroptosis in the MCAO rat model and OGD-treated HT22 cells by modulating the netrin-1/DCC/VEGF/NRF2 axis. In conclusion, our results reveal a novel mechanism for the protection of acute CIs by butylphthalide.
Collapse
Affiliation(s)
- Yuanwei Wang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou
- Department of Neurology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Rui Wei
- Department of Neurology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Li Du
- Department of Neurology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| |
Collapse
|
15
|
Nagase T, Yasuhara T, Kin K, Sasada S, Kawauchi S, Yabuno S, Sugahara C, Hirata Y, Miyake H, Sasaki T, Kawai K, Tanimoto S, Saijo T, Tanaka S. Therapeutic effects of intracerebral transplantation of human modified bone marrow-derived stromal cells (SB623) with voluntary and forced exercise in a rat model of ischemic stroke. Exp Neurol 2025; 386:115145. [PMID: 39805465 DOI: 10.1016/j.expneurol.2025.115145] [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: 09/29/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Ischemic stroke results in significant long-term disability and mortality worldwide. Although existing therapies, such as recombinant tissue plasminogen activator and mechanical thrombectomy, have shown promise, their application is limited by stringent conditions. Mesenchymal stem cell (MSC) transplantation, especially using SB623 cells (modified human bone marrow-derived MSCs), has emerged as a promising alternative, promoting neurogenesis and recovery. This study evaluated the effects of voluntary and forced exercise, alone and in combination with SB623 cell transplantation, on neurological and psychological outcomes in a rat model of ischemic stroke. Male Wistar rats that had undergone middle cerebral artery occlusion (MCAO) were divided into six groups: control, voluntary exercise (V-Ex), forced exercise (F-Ex), SB623 transplantation, SB623 + V-Ex, and SB623 + F-Ex. Voluntary exercise was facilitated using running wheels, while forced exercise was conducted on treadmills. Neurological recovery was assessed using the modified neurological severity score (mNSS). Psychological symptoms were evaluated through the open field test (OFT) and forced swim test (FST), and neurogenesis was assessed via BrdU labeling. Both exercise groups exhibited significant changes in body weight post-MCAO. Both exercises enhanced the treatment effect of SB623 transplantation. The forced exercise showed a stronger treatment effect on ischemic stroke than voluntary exercise alone, and the sole voluntary exercise improved depression-like behavior. The SB623 + F-Ex group demonstrated the greatest improvements in motor function, infarct area reduction, and neurogenesis. The SB623 + V-Ex group was most effective in alleviating depression-like behavior. Future research should optimize these exercise protocols and elucidate the underlying mechanisms to develop tailored rehabilitation strategies for stroke patients.
Collapse
Affiliation(s)
- Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yuichi Hirata
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Hayato Miyake
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tomoya Saijo
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
16
|
Yang Y, Duan Y, Yue J, Yin Y, Ma Y, Wan X, Shao J. Exosomes: an innovative therapeutic target for cerebral ischemia-reperfusion injury. Front Pharmacol 2025; 16:1552500. [PMID: 40206077 PMCID: PMC11979243 DOI: 10.3389/fphar.2025.1552500] [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: 12/28/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Ischemic stroke is caused by artery stenosis or occlusion, which reduces blood flow and may cause brain damage. Treatment includes restoring blood supply; however, ischemia-reperfusion can still aggravate tissue injury. Reperfusion injury can increase levels of reactive oxygen species, exacerbate mitochondrial dysfunction, create excessive autophagy and ferroptosis, and cause inflammation during microglial infiltration. Cerebral ischemia-reperfusion injury (CIRI) is a key challenge in the treatment of ischemic stroke. Currently, thrombolysis (e.g., rt-PA therapy) and mechanical thrombectomy are the primary treatments, but their application is restricted by narrow therapeutic windows (<4.5 h) and risks of hemorrhagic complications. Exosomes reduce CIRI by regulating oxidative stress, mitochondrial autophagy, inflammatory responses, and glial cell polarization. In addition, their noncellular characteristics provide a safer alternative to stem cell therapy. This article reviews the research progress of exosomes in CIRI in recent years.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yushan Duan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jinxi Yue
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yue Yin
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiming Ma
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaohong Wan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jianlin Shao
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
17
|
Li C, Ji H, Mao W, Zhang L, Tong T, Wang J, Cai L, Wang H, Sun T, Yi H, Li S, Tu Y, Zhang J, Wang H, Wu H, Han W, Wang Y. Electroacupuncture regulates FTO/Nrf2/NLRP3 axis-mediated pyroptosis in cerebral ischemia-reperfusion injury. Neuroreport 2025; 36:257-266. [PMID: 39976072 DOI: 10.1097/wnr.0000000000002146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Electroacupuncture (EA) demonstrates neuroprotective in cerebral ischemia-reperfusion (I/R) injury. N6-methyladenosine (m6A) is found to contribute to the pathogenesis of neurological conditions recently. The objective of this study is to investigate the effects of EA on m6A and related mechanism in cerebral I/R injury. After the middle cerebral artery occlusion/reperfusion (MCAO/R) operation was used to establish rat models with cerebral I/R injury, EA was applied to Baihui (GV20) and Dazhui (GV14) once daily for 7 consecutive days. Subsequently, the modified Neurological Severity Score, 2,3,5-triphenyltetrazolium chloride staining, and hematoxylin and eosin staining were performed to assess the neurological damage. To investigate the potential target, the total RNA m6A level and relevant regulators (METTL3, METTL14, WTAP, FTO, and ALKBH5) were examined. In the next step, FTO, Nrf2, NLRP3, IL-18, IL-1β, and TUNEL-positive rates were detected, while the shRNA-FTO was administered to suppress FTO expression. EA improved neurobehavioral disorders, infarct volume, and pathological damage induced by cerebral I/R injury. Mechanically, EA reduced the total RNA m6A level by selectively regulating FTO, but not METTL3, METTL14, WTAP, and ALKBH5. Furthermore, EA could enhance Nrf2 and suppress NLRP3, IL-18, IL-1β, and TUNEL-positive rates, which was reversed by the shRNA-FTO injection. Our findings indicate that EA may alleviate FTO/Nrf2/NLRP3 axis-mediated pyroptosis in cerebral I/R injury, providing a more unified understanding of the neuroprotective effects of EA. Specifically, EA intervention appears to promote the expression of FTO, leading to a reduction of m6A level, which activates Nrf2 and subsequently suppresses NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Chenglong Li
- The First Clinical Medical College (First Affiliated Hospital), Anhui University of Chinese Medicine
| | - Haisheng Ji
- Department of Encephalopathy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province
| | - Wei Mao
- Department of Acupuncture and Moxibustion, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province
| | - Lida Zhang
- Department of Acupuncture and Moxibustion, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province
| | - Tingting Tong
- Department of Encephalopathy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province
| | - Junli Wang
- The First Clinical Medical College (First Affiliated Hospital), Anhui University of Chinese Medicine
| | - Liyuan Cai
- Graduate School, Anhui University of Chinese Medicine, Hefei
| | - Hai Wang
- Graduate School, Anhui University of Chinese Medicine, Hefei
| | - Tingting Sun
- Graduate School, Anhui University of Chinese Medicine, Hefei
| | - Hu Yi
- Graduate School, Anhui University of Chinese Medicine, Hefei
| | - Shijun Li
- Graduate School, Anhui University of Chinese Medicine, Hefei
| | - Ying Tu
- Graduate School, Anhui University of Chinese Medicine, Hefei
| | - Junyu Zhang
- Department of Encephalopathy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province
| | - Haitao Wang
- Department of Encephalopathy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province
| | - Haiyang Wu
- Department of Encephalopathy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province
| | - Wei Han
- The First Clinical Medical College (First Affiliated Hospital), Anhui University of Chinese Medicine
- Department of Acupuncture and Moxibustion, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province
| | - Ying Wang
- The First Clinical Medical College (First Affiliated Hospital), Anhui University of Chinese Medicine
- Department of Encephalopathy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province
- Anhui Huatuo Academy of Traditional Chinese Medicine, Bozhou Vocational and Technical College, Bozhou, Anhui Province, China
| |
Collapse
|
18
|
Song W, Teng L, Wang H, Pang R, Liang R, Zhu L. Exercise preconditioning increases circulating exosome miR-124 expression and alleviates apoptosis in rats with cerebral ischemia-reperfusion injury. Brain Res 2025; 1851:149457. [PMID: 39824375 DOI: 10.1016/j.brainres.2025.149457] [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: 10/05/2024] [Revised: 12/22/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVES Exercise as a non-pharmacological intervention can exert beneficial effects directly through exosomes crossing the blood-brain barrier and reduce apoptosis after cerebral ischaemia/reperfusion injury (CI/RI). miRNA-124 (miR-124) is present in exosomes and plays an important role in regulating cerebral neurological activity; however, the mechanism of the relationship between exercise and the activity of exosomes and apoptosis after CI/RI remains unclear. Therefore, the present study investigated the effects of exercise preconditioning on CI/RI from the perspective of exosomal miR-124 and apoptosis. METHODS The middle cerebral artery occlusion/reperfusion (MCAO/R) model was established by blocking the middle cerebral artery, and a motorized running wheel was chosen as the method of exercise preconditioning for rats, the morphology, particle concentration and particle size distribution of the exosome samples were identified at the 6 h, 12 h, and 24 h time points. RT-PCR, western blotting, immunohistochemistry, TUNEL staining, TTC staining and mNSS scores were used to investigate the effects of exercise preconditioning on apoptosis in MCAO/R rats. RESULTS The results showed exercise reduced neurological dysfunction and infarct size, increased the content of plasma exocrine miR-124 at 24 h, which inhibited the expression of STAT3, increased the expression of the anti-apoptotic BCL-2, and decreased the expression of the pro-apoptotic BAX, thereby reducing apoptosis. CONCLUSIONS Our findings indicated that exercise preconditioning can enhance the anti-apoptotic capacity of tissues in the rat ischemic penumbra and reduce apoptosis after CI/RI via the exosomal miR-124, STAT3, BCL-2/BAX pathway.
Collapse
Affiliation(s)
- Wenjing Song
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Lili Teng
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haoran Wang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Ruifeng Pang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Luwen Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150000, China.
| |
Collapse
|
19
|
Sha Z, Dong S, Nie M, Liu T, Wu C, Lv C, Liu M, Jiang W, Yuan J, Qian Y, Piao X, Jiang R, Gao C. Genetic deletion of G protein-coupled receptor 56 aggravates traumatic brain injury through the microglial CCL3/4/5 upregulation targeted to CCR5. Cell Death Dis 2025; 16:175. [PMID: 40089481 PMCID: PMC11910551 DOI: 10.1038/s41419-025-07501-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 02/10/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Traumatic brain injury (TBI) is a significant global health concern that often results in death or disability, and effective pharmacological treatments are lacking. G protein-coupled receptor 56 (GPR56), a potential drug target, is crucial for neuronal and glial cell function and therefore plays important roles in various neurological diseases. Here, we investigated the potential role and mechanism of GPR56 in TBI-related damage to gain new insights into the pharmacological treatment of TBI. Our study revealed that TBI caused a significant decrease in GPR56 expression and that the deletion of Gpr56 exacerbated neurological function deficits and blood‒brain barrier (BBB) damage following TBI. Additionally, Gpr56 deletion led to increased microgliosis, increased infiltration of peripheral T cells and macrophages, and increased release of cerebral inflammatory cytokines and chemokines after TBI. Furthermore, Gpr56 deletion induced neuronal apoptosis, impaired autophagy, and exacerbated neurological function deficits through microglial-to-neuronal CCR5 signaling after TBI. Overall, these results indicate that Gpr56 knockout exacerbates neurological deficits, neuroinflammation and neuronal apoptosis following TBI through microglial CCL3/4/5 upregulation targeted to CCR5, which indicates that GRP56 may be a potential new pharmacological target for TBI.
Collapse
MESH Headings
- Animals
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/deficiency
- Brain Injuries, Traumatic/genetics
- Brain Injuries, Traumatic/pathology
- Brain Injuries, Traumatic/metabolism
- Microglia/metabolism
- Microglia/pathology
- Receptors, CCR5/metabolism
- Receptors, CCR5/genetics
- Mice
- Up-Regulation
- Chemokine CCL3/metabolism
- Chemokine CCL3/genetics
- Mice, Knockout
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/pathology
- Mice, Inbred C57BL
- Chemokine CCL5/metabolism
- Chemokine CCL5/genetics
- Chemokine CCL4/metabolism
- Chemokine CCL4/genetics
- Male
- Gene Deletion
- Neurons/metabolism
- Neurons/pathology
- Apoptosis
- Signal Transduction
Collapse
Affiliation(s)
- Zhuang Sha
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Shiying Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Meng Nie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Tao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Chenrui Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Chuanxiang Lv
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Mingqi Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Weiwei Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Jiangyuan Yuan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Yu Qian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Xianhua Piao
- Weill Institute for Neuroscience, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Newborn Brain Research Institute, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Rongcai Jiang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- State Key Laboratory of Experimental Hematology, Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Chuang Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.
| |
Collapse
|
20
|
Du J, Liang X, Wang D, Wang Z, Shen R. Mechanism of KDM4A in Regulating Microglial Polarization in Ischemic Stroke. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05207-2. [PMID: 40080374 DOI: 10.1007/s12010-025-05207-2] [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] [Accepted: 12/24/2024] [Indexed: 03/15/2025]
Abstract
Microglia polarization plays important roles in inflammatory processes after ischemic stroke. This study aimed to explore the mechanism of lysine-specific histone demethylase 4 (KDM4A) in microglia polarization after ischemic stroke. The mouse model was established using middle cerebral artery occlusion/reperfusion (MCAO/R) and the cell model was established by oxygen-glucose deprivation/reperfusion (OGD/R). The neurological deficits and brain tissue injury were evaluated. The biomarkers of microglia were determined. Levels of KDM4A/mouse double minute-2 homolog (MDM2)/C1q/TNF-related protein-3 (CTRP3) were measured. Inflammatory cytokines were quantified. The impact of KDM4A on microglia polarization was assessed. The enrichment of KDM4A or histone 3 lysine 9 trimethylation (H3K9me3) on the MDM2 promoter was analyzed. The ubiquitination and protein levels of CTRP3 after MG132 and cycloheximide treatment were determined. Results showed that KDM4A and MDM2 were upregulated while CTRP3 was downregulated. KDM4A downregulation alleviated neurological dysfunction, rescued motor capacity, reduced inflammatory infiltration, suppressed microglia activation, and promoted M2 polarization. KDM4A inhibited the enrichment of H3K9me3 on the MDM2 promoter, increasing MDM2 expression and downregulating CTRP3 expression via ubiquitination and degradation. MDM2 overexpression or CTRP3 downregulation averted the promotive role of silencing KDM4A in microglia polarization. In conclusion, KDM4A promotes microglia polarization to aggravate ischemic stroke via the MDM2/CTRP3 axis.
Collapse
Affiliation(s)
- Jingliang Du
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Xianyang Liang
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Denghui Wang
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Ruile Shen
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China.
| |
Collapse
|
21
|
Challa SR, Levingston H, Fornal CA, Baker IM, Boston J, Shanthappa N, Unnam P, Klopfenstein JD, Veeravalli KK. Temporal mRNA Expression of Purinergic P2 Receptors in the Brain Following Cerebral Ischemia and Reperfusion: Similarities and Distinct Variations Between Rats and Mice. Int J Mol Sci 2025; 26:2379. [PMID: 40141023 PMCID: PMC11941906 DOI: 10.3390/ijms26062379] [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: 01/23/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Purinergic P2 receptors are crucial in energy utilization and cellular signaling, making them key targets for stroke therapies. This study examines the temporal mRNA expression of all P2 receptors in rats and mice. Both species exhibited a common subset of P2X and P2Y receptors with elevated expression following cerebral ischemia and reperfusion (I/R), highlighting conserved mechanisms across these species. The receptors with upregulated expression in both species were P2X3, P2X4, P2X7, P2Y2, and P2Y6. While these similarities were observed, notable differences in receptor expression emerged between rats and mice. Rats exhibited a broader receptor profile, with five additional receptors (P2X1, P2Y1, P2Y12, P2Y13, and P2Y14) significantly upregulated compared to only two receptors (P2X2 and P2Y4) in mice, highlighting species-specific regulation of receptor expression distinct from the shared receptors. Following cerebral I/R, P2Y12 was the most upregulated receptor in rats, while P2Y2 was the most upregulated in mice. These findings reveal both conserved and species-specific changes in P2 receptor expression following cerebral I/R. Targeting purinergic receptors, particularly those conserved and upregulated in response to stroke, may represent a promising therapeutic approach.
Collapse
Affiliation(s)
- Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Hunter Levingston
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Casimir A. Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Isidra M. Baker
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Joseph Boston
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Nidhi Shanthappa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Pavani Unnam
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Jeffrey D. Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Illinois Neurological Institute, OSF HealthCare, Peoria, IL 61603, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Neurology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| |
Collapse
|
22
|
Kim D, Lee JW, Kim YT, Choe J, Kim G, Ha CM, Kim JG, Song KH, Yang S. Minimally Invasive Syringe-Injectable Hydrogel with Angiogenic Factors for Ischemic Stroke Treatment. Adv Healthc Mater 2025; 14:e2403119. [PMID: 39520382 PMCID: PMC11874675 DOI: 10.1002/adhm.202403119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/06/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke (IS) accounts for most stroke incidents and causes intractable damage to brain tissue. This condition manifests as diverse aftereffects, such as motor impairment, emotional disturbances, and dementia. However, a fundamental approach to curing IS remains unclear. This study proposes a novel approach for treating IS by employing minimally invasive and injectable jammed gelatin-norbornene nanofibrous hydrogels (GNF) infused with growth factors (GFs). The developed GNF/GF hydrogels are administered to the motor cortex of a rat IS model to evaluate their therapeutic effects on IS-induced motor dysfunction. GNFs mimic a natural fibrous extracellular matrix architecture and can be precisely injected into a targeted brain area. The syringe-injectable jammed nanofibrous hydrogel system increased angiogenesis, inflammation, and sensorimotor function in the IS-affected brain. For clinical applications, the biocompatible GNF hydrogel has the potential to efficiently load disease-specific drugs, enabling targeted therapy for treating a wide range of neurological diseases.
Collapse
Affiliation(s)
- Donggue Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Ji Woo Lee
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Yang Tae Kim
- Division of Life SciencesCollege of Life Sciences and BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Junhyeok Choe
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Gaeun Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Chang Man Ha
- Research Division and Brain Research Core Facilities of Korea Brain Research InstituteDaegu41068Republic of Korea
| | - Jae Geun Kim
- Division of Life SciencesCollege of Life Sciences and BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
| | - Kwang Hoon Song
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
| | - Sunggu Yang
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
- gBrain Inc.Incheon21984Republic of Korea
| |
Collapse
|
23
|
Lee Y, Lee J, Kim J, Cho S, Lee HJ, Geum D, Park DH, Kim JH. hESC-derived extracellular vesicles enriched with MFGE-8 and the GSH redox system act as senotherapeutics for neural stem cells in ischemic stroke. Free Radic Biol Med 2025; 229:333-349. [PMID: 39870225 DOI: 10.1016/j.freeradbiomed.2025.01.050] [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: 12/17/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Human embryonic stem cells (hESCs) and their extracellular vesicles (EVs) hold significant potential for tissue repair and regeneration. Neural stem cells (NSCs) in the adult brain often acquire senescent phenotypes after ischemic injuries, releasing neurodegenerative senescence-associated secretory phenotype factors. In this study, we investigated the senotherapeutic effects of hESC-EVs on NSCs and confirmed their neuroprotective effects in neurons via rejuvenation of NSC secretions. Proteomic profiling of hESC-EVs identified MFGE-8 as a critical bridging molecule to NSCs. We also found that the glutathione (GSH) redox system is a key contributor to the therapeutic antioxidant activity of hESC-EVs. Additionally, EVs produced by the hypoxic preconditioning of hESCs (hESC-HypoxEVs) exhibited reinforced GSH redox capacity and further enhanced the senotherapeutic effects on NSCs compared to naïve hESC-EVs. We also demonstrated that administration of hESC-HypoxEVs, precoated with MFGE-8, significantly increased the populations of NSCs and newborn neurons in the subventricular zone of the brain and improved sensorimotor functions in a rat model of ischemic stroke. Our study suggests that combining hESC-HypoxEVs with MFGE-8 may serve as an effective therapeutic modality for reversing senescence and enhancing the neurogenic potential of NSCs to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Youngseok Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea; Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jihun Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jeongjun Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Seunghyun Cho
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Hye-Jin Lee
- Department of Neurosurgery, Anam Hospital, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Dongho Geum
- Department of Convergence Medicine, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Dong-Hyuk Park
- Department of Neurosurgery, Anam Hospital, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
24
|
Sağlam-Çifci E, Güleç İ, Şengelen A, Karagöz-Güzey F, Eren B, Paşaoğlu HE, Önay-Uçar E. The H 4R antagonist, JNJ-7777120 treatments ameliorate mild traumatic brain injury by reducing oxidative damage, inflammatory and apoptotic responses through blockage of the ERK1/2/NF-κB pathway in a rat model. Exp Neurol 2025; 385:115133. [PMID: 39732275 DOI: 10.1016/j.expneurol.2024.115133] [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: 10/26/2024] [Revised: 12/15/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Growing evidence reveals that microglia activation and neuroinflammatory responses trigger cell loss in the brain. Histamine is a critical neurotransmitter and promotes inflammatory responses; thus, the histaminergic system is a potential target for treating neurodegenerative processes. JNJ-7777120, a histamine H4 receptor (H4R) antagonist, has been shown to alleviate inflammation, brain damage, and behavioral deficits effectively, but there is no report on its role in brain trauma. Herein, we investigated the neuroprotective effects of JNJ-7777120 (shortly JNJ) in a mild traumatic brain injury (mTBI). mTBI setup was performed using a weight-drop model in adult male Sprague-Dawley rats. JNJ (1 mg/kg, twice/day for 7 days) was intraperitoneally administered following mTBI. Modified neurological severity score and beam-walking test used to assess motor, sensory, reflex, and balance functions (post-TBI days-1/3/7) showed that JNJ had significantly improved these functions. HE-staining revealed reduced neurodegenerative cells after JNJ-treatments compared to vehicle (2.85 % DMSO) treated group. JNJ also decreased the injury-induced apoptosis (Bax/Bcl-2, cleaved-Cas-3, cleaved-PARP1), oxidative (4HNE, MDA), and inflammatory (IBA1, TNF-α, IL-1β, IL-6, and IL-10) responses. Furthermore, blocking the activation of the ERK1/2/NF-κB pathway was determined to be involved in its therapeutic mechanism. The network pharmacology analyses for JNJ-7777120 and TBI confirmed the importance of targeting neurotransmitter receptor activity, signaling receptor activity, and kinase activation. Our results provide the first proof of the efficacy of an H4R antagonist in a mild TBI rat model and suggest that H4R targeting by JNJ-treatment might be a promising therapeutic approach to clinically halt the progression of brain injury.
Collapse
Affiliation(s)
- Ece Sağlam-Çifci
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - İlker Güleç
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye.
| | - Feyza Karagöz-Güzey
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Burak Eren
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Hüsniye Esra Paşaoğlu
- Department of Pathology, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Evren Önay-Uçar
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye.
| |
Collapse
|
25
|
Yang X, Zhao Y, Yu S, Chi L, Cai Y. Coenzyme Q10 alleviates neurological deficits in a mouse model of intracerebral hemorrhage by reducing inflammation and apoptosis. Exp Biol Med (Maywood) 2025; 250:10321. [PMID: 40093659 PMCID: PMC11906280 DOI: 10.3389/ebm.2025.10321] [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: 07/19/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
This research study was directed towards to assessing whether coenzyme Q10 (CoQ10) is linked to neuroprotection and induces anti-inflammatory and anti-neuronal death responses in an Intracerebral hemorrhage (ICH) mouse model via right caudate nucleus injection with collagenase VII. Autologous blood was injected into mice to induce ICH. We found that FoxM1 was upregulated in the ICH-injured animals. Moreover, CoQ10 treatment effectively ameliorated neurological deficits, mitigated cerebral edema, and minimized hematoma in model mice, demonstrating dose-dependent efficacy and promoting the functional recovery of the animals. ELISA and real-time PCR assays of pro-inflammatory cytokines indicated that CoQ10 was capable of alleviating neuroinflammation in ICH. In line with the part of CoQ10 in attenuating the inflammatory response, CoQ10 also suppressed cell apoptosis in the ICH-injured brain, which partly accounts for its neuroprotective effect. Furthermore, our analysis of different inflammatory pathways indicated that CoQ10 targeted the nuclear factor-kappa B signaling axis. Our findings suggest that CoQ10 protects against ICH by mitigating neuroinflammatory responses and preventing neuronal apoptosis, with the underlying mechanism possibly being connected with nuclear factor-kappa B pathway regulation. Therefore, CoQ10 holds significant potential as a therapeutic strategy for treating ICH.
Collapse
Affiliation(s)
- Xiaoqing Yang
- Department of Neurosurgery, Ruian People’s Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Zhao
- Department of Traditional Chinese Medicine, Ruian Tangxia People’s Hospital, Wenzhou, Zhejiang, China
| | - Sisi Yu
- Department of Neurosurgery, Ruian People’s Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lihui Chi
- Department of Neurosurgery, Ruian People’s Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yeyan Cai
- Department of Neurosurgery, Ruian People’s Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
26
|
Rolland TJ, Zahra S, Cucinotta D, Young R, Weil B. Mesenchymal Stem Cell-Derived Extracellular Vesicles Mitigate Immune Cell Activation in an In Vitro Model of Post-Resuscitation Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.637856. [PMID: 40027652 PMCID: PMC11870425 DOI: 10.1101/2025.02.13.637856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Systemic inflammation is a well-established component of post-cardiac arrest syndrome (PCAS), a condition responsible for significant morbidity and mortality in patients who are initially resuscitated from sudden cardiac arrest. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as promising immunomodulatory agents in various inflammatory conditions, including after ischemia-reperfusion injury (IRI). Here, we investigated the therapeutic potential of MSC-EVs in porcine peripheral blood mononuclear cells (PBMCs) stimulated with lipopolysaccharide (LPS) or mitochondrial DNA (mtDNA) to mimic immune cell activation in PCAS. Methods PBMCs were isolated from healthy pigs ( Sus scrofa ), cultured in vitro , stimulated with LPS or mtDNA, and treated with a range of MSC-EV concentrations. Flow cytometry, quantitative PCR, ELISA, and ROS/RNS measurements were performed to assess PBMC activation. Results MSC-EV treatment reduced LPS-induced inflammatory granulocyte activation and selectively modulated cytokine transcripts, including IFNα, IL-1β, and TNF-α, in a concentration-dependent manner. Similar immunosuppressive effects were observed in mtDNA-stimulated PBMCs, where MSC-EVs attenuated dendritic cell activation and inflammatory cytokine release. Furthermore, higher concentrations of MSC-EVs significantly decreased ROS/RNS production in both LPS- and mtDNA-challenged PBMCs. Conclusions MSC-EVs exhibit potent immunomodulatory properties against LPS- and mtDNA-induced activation of porcine PBMCs, highlighting their broad capacity to modulate immune responses and mitigate oxidative stress induced by pro-inflammatory stimuli that are relevant to PCAS. These findings provide further support for the administration of MSCs, or MSC-EVs themselves, as a potential therapeutic intervention to target immune activation in PCAS and other disorders characterized by an acute systemic inflammatory state.
Collapse
|
27
|
Bayat Tork MA, Saberifar M, Joneidi Yekta H, Hajinejad M, Hosseini Ravandi H, Gorji A, Sahab Negah S. Nano-scaffold containing functional motif of stromal cell-derived factor 1 enhances neural stem cell behavior and synaptogenesis in traumatic brain injury. Sci Rep 2025; 15:5811. [PMID: 39962142 PMCID: PMC11832925 DOI: 10.1038/s41598-025-85698-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and morbidity worldwide, presenting a significant challenge due to the lack of effective therapies. Neural stem cells (NSCs) have shown promising potential in preclinical studies as a therapy for TBI. However, their application is limited by challenges related to poor survival and integration within the injured brain. This study investigated the effect of a novel nano-scaffold containing stromal cell-derived factor 1 (SDF-1) on NSC behavior and synaptogenesis after TBI. Using an innovative design, we successfully fabricated a nano-scaffold with Young's modulus of approximately 3.21 kPa, which aligns closely with the mechanical properties exhibited by neural tissue. This achievement marks the first time such a scaffold has been created and has promising implications for its potential use in neural tissue engineering applications. Our findings demonstrate that the nano-scaffold enhances NSC proliferation, migration, and differentiation capacity in vitro. Moreover, when transplanted into the injured brain, the nano-scaffold promotes the survival and integration of NSCs, leading to increased synaptogenesis and functional recovery. These findings suggest that using the novel nano-scaffold containing SDF-1 could provide a promising approach to treating TBI by improving NSC behavior and promoting synaptogenesis.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Saberifar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Joneidi Yekta
- New Technologies Research Center, Amirkabir University of Technology, Tehran, Iran
| | - Mehrdad Hajinejad
- Qaen Faculty of Medical Science, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery, Westfälische Wilhelms-Universität, 48149, Munster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, 48149, Munster, Germany.
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Huang H, Jiang NN, Lu GW, Xu F, Sun LL, Zhu J, Dong Z, Zhang ZJ, Liu S. CircMETTL9 targets CCAR2 to induce neuronal oxidative stress and apoptosis via mitochondria-mediated pathways following traumatic brain injury. Free Radic Biol Med 2025; 228:44-61. [PMID: 39709098 DOI: 10.1016/j.freeradbiomed.2024.12.041] [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: 10/01/2024] [Revised: 11/18/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
Traumatic brain injury (TBI) remains a principal factor in neurological disorders, often resulting in significant morbidity due to secondary neuroinflammatory and oxidative stress responses. While circular RNAs are recognized for their high expression levels in the nervous system and play crucial roles in various neurological processes, their specific contributions to the pathophysiology of TBI remain underexplored. In this study, the possible molecular mechanisms through which circMETTL9 modulated oxidative stress and neurological outcomes following TBI were investigated. In vitro model of oxidative stress utilizing SH-SY5Y cells revealed that circMETTL9 knockdown significantly attenuated H₂O₂-induced reactive oxygen species (ROS) production, reduced apoptosis, and preserved mitochondrial function. Additionally, CCAR2 has been identified as a circMETTL9-binding protein by mass spectrometry and RNA immunoprecipitation, with circMETTL9 positively regulating CCAR2 expression. Meanwhile, on the basis of silencing CCAR2, it was verified that the regulation of oxidative stress in SH-SY5Y cells by circMETTL9 was mediated by CCAR2. In vivo experiments using a TBI rat model further confirmed that CCAR2 knockdown alleviated central nervous system (CNS) injury, reduced oxidative stress and apoptosis, and protected mitochondrial integrity following TBI. These findings suggest a novel mechanism by which circMETTL9 targets CCAR2 via mitochondria-mediated Bax/Bcl-2/caspase-3 signaling to regulate apoptosis. CircMETTL9 may provide a viable therapeutic target for mitigating neurological dysfunction following TBI, offering new insights into potential interventions aimed at reducing secondary brain injury.
Collapse
Affiliation(s)
- Hao Huang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; School of Nursing and Rehabilitation, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Nan-Nan Jiang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; School of Nursing and Rehabilitation, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Gui-Wei Lu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; School of Nursing and Rehabilitation, Nantong University, Nantong, 226001, Jiangsu Province, China; Department of Rehabilitation Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu Province, China
| | - Feng Xu
- The Second People's Hospital of Nantong, Nantong, 226002, Jiangsu Province, China
| | - Lu-Lu Sun
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Jing Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Zhao Dong
- Nanjing Vocational Health College, Nanjing, 210038, Jiangsu Province, China.
| | - Zhi-Jun Zhang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, 226001, Jiangsu Province, China.
| | - Su Liu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
29
|
Feng S, Li J, Liu T, Huang S, Chen X, Liu S, Zhou J, Zhao H, Hong Y. Overexpression of low-density lipoprotein receptor prevents neurotoxic polarization of astrocytes via inhibiting NLRP3 inflammasome activation in experimental ischemic stroke. Neural Regen Res 2025; 20:491-502. [PMID: 38819062 PMCID: PMC11317962 DOI: 10.4103/nrr.nrr-d-23-01263] [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: 07/28/2023] [Revised: 12/26/2023] [Accepted: 02/23/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00027/figure1/v/2024-05-28T214302Z/r/image-tiff Neurotoxic astrocytes are a promising therapeutic target for the attenuation of cerebral ischemia/reperfusion injury. Low-density lipoprotein receptor, a classic cholesterol regulatory receptor, has been found to inhibit NLR family pyrin domain containing protein 3 (NLRP3) inflammasome activation in neurons following ischemic stroke and to suppress the activation of microglia and astrocytes in individuals with Alzheimer's disease. However, little is known about the effects of low-density lipoprotein receptor on astrocytic activation in ischemic stroke. To address this issue in the present study, we examined the mechanisms by which low-density lipoprotein receptor regulates astrocytic polarization in ischemic stroke models. First, we examined low-density lipoprotein receptor expression in astrocytes via immunofluorescence staining and western blotting analysis. We observed significant downregulation of low-density lipoprotein receptor following middle cerebral artery occlusion reperfusion and oxygen-glucose deprivation/reoxygenation. Second, we induced the astrocyte-specific overexpression of low-density lipoprotein receptor using astrocyte-specific adeno-associated virus. Low-density lipoprotein receptor overexpression in astrocytes improved neurological outcomes in middle cerebral artery occlusion mice and reversed neurotoxic astrocytes to create a neuroprotective phenotype. Finally, we found that the overexpression of low-density lipoprotein receptor inhibited NLRP3 inflammasome activation in oxygen-glucose deprivation/reoxygenation injured astrocytes and that the addition of nigericin, an NLRP3 agonist, restored the neurotoxic astrocyte phenotype. These findings suggest that low-density lipoprotein receptor could inhibit the NLRP3-meidiated neurotoxic polarization of astrocytes and that increasing low-density lipoprotein receptor in astrocytes might represent a novel strategy for treating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Shuai Feng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Juanji Li
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tingting Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shiqi Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shen Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongdong Zhao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
30
|
Shi D, Bai Y, Long R, Xia J, Xu W, Qin D, Yang X, Ding M, Hou XY. Neuronal LAMP2A-mediated reduction of adenylyl cyclases induces acute neurodegenerative responses and neuroinflammation after ischemic stroke. Cell Death Differ 2025; 32:337-352. [PMID: 39341961 PMCID: PMC11802923 DOI: 10.1038/s41418-024-01389-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Lysosomes regulate cellular metabolism to maintain cell survival, but the mechanisms whereby they determine neuronal cell fate after acute metabolic stress are unknown. Neuron-enriched lysosomal membrane protein LAMP2A is involved in selective chaperone-mediated autophagy and exosome loading. This study demonstrates that abnormalities in the neuronal LAMP2A-lysosomal pathway cause neurological deficits following ischemic stroke and that this is an early inducer of the PANoptosis-like molecular pathway and neuroinflammation, simultaneously inducing upregulation of FADD, RIPK3, and MLKL after ischemia. Quantitative proteomic and pharmacological analysis showed that after acute metabolic stress, the neuronal LAMP2A pathway induced acute synaptic degeneration and PANoptosis-like responses involving downregulation of protein kinase A (PKA) signaling. LAMP2A directed post-stroke lysosomal degradation of adenylyl cyclases (ADCY), including ADCY1 and ADCY3 in cortical neurons. Post-stroke treatment with cAMP mimetic or ADCY activator salvaged cortical neurons from PANoptosis-like responses and neuroinflammation, suggesting that the neuronal ADCY-cAMP-PKA axis is an upstream arrester of the pathophysiological process following an ischemic stroke. This study demonstrates that the neuronal LAMP2A-lysosmal pathway drives intricate acute neurodegenerative and neuroinflammatory responses after brain metabolic stress by downregulating the ADCY-PKA signaling cascade, and highlights the therapeutic potential of PKA signal inducers for improving stroke outcomes.
Collapse
Affiliation(s)
- Dingfang Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yunhao Bai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ruiling Long
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jing Xia
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenxuan Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Dongshen Qin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xuejun Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ming Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Xiao-Yu Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
31
|
Liu J, Li M, Huang Y, Wang X, Xu Y, Fu Z, Lin Z, Chen J, Wu X. Inclusion Complex of Nimodipine with Sulfobutylether-β-cyclodextrin: Preparation, Characterization, In Vitro and In Vivo Evaluation. AAPS PharmSciTech 2025; 26:28. [PMID: 39779582 DOI: 10.1208/s12249-024-03014-2] [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: 08/19/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Nimodipine (NIMO) is used to treat ischemic nerve injury from subarachnoid hemorrhage (SAH), but its low aqueous solubility limits clinical safety and bioavailability. This study aims to improve NIMO's solubility by preparing inclusion complexes with sulfobutylether-β-cyclodextrin (SBE-β-CD), reducing the limitations of Nimotop® injection, including vascular irritation, toxicity, and poor dilution stability. The NIMO-SBE-β-CD inclusion complex (NIMO-CD) was characterized in both liquid and solid states through phase solubility studies and methods including DSC, FT-IR, XRD, and SEM. Dilution stability, hemolysis, vascular irritation, and acute toxicity tests were performed, with pharmacokinetic and pharmacodynamic studies using Nimotop® as the control. Physical characterization confirmed the successful formation of the inclusion complex. NIMO's solubility improved by 1202-fold (from 0.82 to 986.19 μg/mL at 25℃). NIMO-CD showed stability for 24 h when diluted, exhibited no hemolytic activity, reduced vascular irritation, and its median lethal dose (LD50) was 2.49 times higher than that of Nimotop®. Both NIMO-CD and Nimotop® displayed similar pharmacokinetic profiles. Behavioral assessments (mNSS scoring and CT), along with evaluations of hematoma area and histopathology, demonstrated that NIMO-CD significantly improved outcomes in intracerebral hemorrhage, greatly enhancing neurological recovery, reducing hematoma and edema, and achieving treatment effects comparable to those of Nimotop® injection. NIMO-CD significantly improves NIMO's solubility and stability while maintaining bioequivalence with Nimotop®. Furthermore, its enhanced safety profile indicates its potential as a superior formulation for treating ischemic nerve injuries.
Collapse
Affiliation(s)
- Jiahui Liu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
- Shanghai Wei Er Lab, Shanghai, China
| | - Meichai Li
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
- Shanghai Wei Er Lab, Shanghai, China
| | - Yongjie Huang
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | | | - Youfa Xu
- Shanghai Wei Er Lab, Shanghai, China
| | - Zhiqin Fu
- Shanghai Wei Er Lab, Shanghai, China
| | | | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China.
- Shanghai Wei Er Lab, Shanghai, China.
| | - Xin Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China.
- Shanghai Wei Er Lab, Shanghai, China.
| |
Collapse
|
32
|
Liu Y, Yixilamu, Jin G, Feng M, Chunhua, Dawa. Tibetan golden acupuncture inhibits JNK/caspase-3 signaling pathway to alleviate neuronal apoptosis in cerebral ischemia-reperfusion injury. Heliyon 2024; 10:e40443. [PMID: 39698073 PMCID: PMC11652837 DOI: 10.1016/j.heliyon.2024.e40443] [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: 02/06/2024] [Revised: 07/17/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Background Apoptosis induced by cerebral ischemia-reperfusion is one of the key pathological processes of nerve injury. Tibetan golden acupuncture (GA) is a common treatment for ischemic brain injury in Tibetan. The aim of this study was to explore whether GA prevents cerebral ischemia-reperfusion-induced apoptosis in mice by blocking the JNK/caspase-3 pathway. Methods In experiment I, 36 mice were randomly divided into a Sham group, CI/RI group, CI/RI + GA group. Morris water maze tests, TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining and flow cytometry (FCM) were used to evaluate the effect of the GA intervention on CI/RI. In experiment II, 30 mice were randomly divided into a Sham group, CI/RI group, CI/RI + GA group, CI/RI + SP group and CI/RI + SP + EA group. Western blotting was used to detect protein expression of key factors in the JNK signaling pathway in the hippocampus. Results After 7 and 14 interventions, behavioral evaluations in CI/RI + GA group was significantly different from those in CI/RI groups (p < 0.01), pathological injury and apoptosis were significantly reduced (p < 0.01). Compared with CI/RI group, the expression of P-JNK/JNK, Cleaved caspase-3/caspase-3, Bax, and Bad proteins in CI/RI + GA group, CI/RI + SP and CI/RI + SP + GA groups were significantly decreased (p < 0.01). The expression of B-cell lymphoma 2 (Bcl-2) was significantly increased (p < 0.01, p < 0.05). Conclusions GA can restore neurological dysfunction and inhibit hippocampal neuronal apoptosis in CI/RI mice, at least partially through inhibition of the JNK/Caspase-3 signaling pathway and regulation of apoptosis signals.
Collapse
Affiliation(s)
- Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
- University of Tibetan Medicine, Lhasa, China
| | - Yixilamu
- University of Tibetan Medicine, Lhasa, China
| | - Guilin Jin
- University of Tibetan Medicine, Lhasa, China
| | - Mingke Feng
- University of Tibetan Medicine, Lhasa, China
| | - Chunhua
- University of Tibetan Medicine, Lhasa, China
| | - Dawa
- University of Tibetan Medicine, Lhasa, China
| |
Collapse
|
33
|
Yi L, Qu Y, Zhang Q, Shi S, Li F, Qu C, Tang Y, Wen S, Pan Y. Enforced hematopoietic cell E-selectin/L-selectin ligand expression enhances bone marrow stromal cells homing and amelioration of cerebral ischemia-reperfusion injury via induction of prostaglandin E2. Stem Cells 2024; 42:1070-1084. [PMID: 39364762 DOI: 10.1093/stmcls/sxae062] [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: 11/17/2023] [Accepted: 08/09/2024] [Indexed: 10/05/2024]
Abstract
Ischemic stroke (IS) is a significant and potentially life-threatening disease with limited treatment options, often resulting in severe disability. Bone marrow stromal cells (BMSCs) transplantation has exhibited promising neuroprotection following cerebral ischemia-reperfusion injury (CIRI). However, the effectiveness is hindered by their low homing rate when administered through the vein. In this study, we aimed to enhance the homing ability of BMSCs through lentivirus transfection to express fucosyltransferase 7. This glycosylation engineered CD44 on BMSCs to express hematopoietic cell E-selectin/L-selectin ligand (HCELL), which is the most potent E-selectin ligand. Following enforced HCELL expression, the transplantation of BMSCs was then evaluated in a middle cerebral artery occlusion model. Results showed that HCELL+BMSCs significantly ameliorated neurological deficits and reduced the volume of cerebral infarction. Furthermore, the transplantation led to a decrease in apoptosis by upregulating BCL-2 and downregulating BAX, also reduced the mRNA levels of inflammatory factors, such as interleukin-1β (IL-1β), IL-2, IL-6, and tumor necrosis factor-alpha (TNF-α) in the ischemic brain tissue. Notably, enforced HCELL expression facilitated the migration of BMSCs toward cerebral ischemic lesions and their subsequent transendothelial migration through the upregulation of PTGS-2, increased production of PGE2 and activation of VLA-4. In summary, our study demonstrates that transplantation of HCELL+BMSCs effectively alleviates CIRI, and that enforced HCELL expression enhances the homing of BMSCs to cerebral ischemic lesions and their transendothelial migration via PTGS-2/PGE2/VLA-4. These findings indicate that enforced expression of HCELL on BMSCs could serve as a promising therapeutic strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lian Yi
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Yewei Qu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Qi Zhang
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin 150001, People's Republic of China
| | - Shanshan Shi
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Fangqin Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Changda Qu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Yushi Tang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Shirong Wen
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Yujun Pan
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| |
Collapse
|
34
|
Bai S, Zhang G, Chen S, Wu X, Li J, Wang J, Chen D, Liu X, Wang J, Li Y, Tang Y, Tang Z. MicroRNA-451 Regulates Angiogenesis in Intracerebral Hemorrhage by Targeting Macrophage Migration Inhibitory Factor. Mol Neurobiol 2024; 61:10481-10499. [PMID: 38743209 PMCID: PMC11584486 DOI: 10.1007/s12035-024-04207-3] [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: 05/31/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke with the highest fatality and disability rate. Up to now, commonly used first-line therapies have limited value in improving prognosis. Angiogenesis is essential to neurological recovery after ICH. Recent studies have shown that microRNA-451(miR-451) plays an important role in angiogenesis by regulating the function of vascular endothelial cells. We found miR-451 was significantly decreased in the peripheral blood of ICH patients in the acute stage. Based on the clinical findings, we conducted this study to investigate the potential regulatory effect of miR-451 on angiogenesis after ICH. The expression of miR-451 in ICH mouse model and in a hemin toxicity model of human brain microvascular endothelial cells (hBMECs) was decreased the same as in ICH patients. MiR-451 negatively regulated the proliferation, migration, and tube formation of hBMECs in vitro. MiR-451 negatively regulated the microvessel density in the perihematoma tissue and affected neural functional recovery of ICH mouse model. Knockdown of miR-451 could recovered tight junction and protect the integrity of blood-brain barrier after ICH. Based on bioinformatic programs, macrophage migration inhibitory factor (MIF) was predicted to be the target gene and identified to be regulated by miR-451 inhibiting the protein translation. And p-AKT and p-ERK were verified to be downstream of MIF in angiogenesis. These results all suggest that miR-451 will be a potential target for regulating angiogenesis in ICH.
Collapse
Affiliation(s)
- Shuang Bai
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingxuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
35
|
Li H, Wei J, Li M, Li Y, Zhang T, Tian J, Liu X, Li K, Lin J. Biological characteristics of Muse cells derived from MenSCs and their application in acute liver injury and intracerebral hemorrhage diseases. Regen Ther 2024; 27:48-62. [PMID: 38496012 PMCID: PMC10940801 DOI: 10.1016/j.reth.2024.03.003] [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: 11/28/2023] [Revised: 02/16/2024] [Accepted: 03/03/2024] [Indexed: 03/19/2024] Open
Abstract
The increasing interest in multilineage differentiating stress-enduring (Muse) cells within the field of regenerative medicine is attributed to their exceptional homing capabilities, prolonged viability in adverse conditions, and enhanced three-germ-layer differentiate ability, surpassing their parent mesenchymal stem cells. Given their abundant sources, non-invasive collection procedure, and periodic availability, human menstrual blood-derived endometrium stem cells (MenSCs) have been extensively investigated as a potential resource for stem cell-based therapies. However, there is no established modality to isolate Muse cells from MenSCs and disparity in gene expression profiles between Muse cells and MenSCs remain unknown. In this study, Muse cells were isolated from MenSCs by long-time trypsin incubation method. Muse cells expressed pluripotency markers and could realize multilineage differentiation in vitro. Compared with MenSCs, Muse cells showed enhanced homing ability and superior therapeutic efficacy in animal models of acute liver injury (ALI) and intracerebral hemorrhage (ICH). Furthermore, the RNA-seq analysis offers insights into the mechanism underlying the disparity in trypsin resistance and migration ability between Muse and MenSCs cells. This research offers a significant foundation for further exploration of cell-based therapies using MenSCs-derived Muse cells in the context of various human diseases, highlighting their promising application in the field of regenerative medicine.
Collapse
Affiliation(s)
- Han Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jinghui Wei
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Mingzhi Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yaoqiang Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Tong Zhang
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jialu Tian
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xuejia Liu
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Kangjia Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
36
|
Ji Q, Liu M, Gao L, Zhang S, Zhang W, Wang M, Xia Z, Li B, Kong L, Yao Y, Wang Y, Li J, Yan Q, Wu S, Liu H, Hu S. Combination of trimethoprim-sulfamethoxazole and mesenchymal stem cell therapy to treat toxoplasmic encephalitis after hematopoietic stem cell transplantation: A case report. Transpl Immunol 2024; 87:102130. [PMID: 39278332 DOI: 10.1016/j.trim.2024.102130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Toxoplasmosis, caused by the parasite Toxoplasma gondii, is a life-threatening infection that may occur following hematopoietic stem cell transplantation (HSCT). Toxoplasmic encephalitis (TE) is one of the most severe manifestations of this infection and often results in unsatisfactory therapeutic outcomes, especially regarding neurological damage. Recent studies have demonstrated that human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) can significantly aid in neural repair and remodeling. Furthermore, hUC-MSCs have been shown to reduce the risk of graft-versus-host disease (GVHD) associated with the reduction or discontinuation of immunosuppressive therapy. In this case report, we present a pediatric patient who developed TE as a complication of haploidentical HSCT. The patient received a combined treatment regimen of standard anti-Toxoplasma therapy and adjunctive hUC-MSC therapy. The outcomes were satisfactory. The patient regained consciousness, maintained a stable body temperature, and regained the ability to perform daily activities independently. Additionally, next-generation sequencing revealed a decrease in Toxoplasma DNA sequences in the blood and cerebrospinal fluid to undetectable levels. This case report underscores the potential of hUC-MSCs as a promising therapeutic modality for TE.
Collapse
Affiliation(s)
- Qi Ji
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Minyuan Liu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Li Gao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Senlin Zhang
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Weiliang Zhang
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Manli Wang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Zihao Xia
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Bohan Li
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Lingjun Kong
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Yanhua Yao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Yi Wang
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Jie Li
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Qing Yan
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Shuiyan Wu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Hu Liu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Shaoyan Hu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Jiangsu Pediatric Hematol & Oncol Center, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China.
| |
Collapse
|
37
|
Gu F, Wang Z, Ding H, Tao X, Zhang J, Dai K, Li X, Shen H, Li H, Chen Z, Wang Z. Microglial mitochondrial DNA release contributes to neuroinflammation after intracerebral hemorrhage through activating AIM2 inflammasome. Exp Neurol 2024; 382:114950. [PMID: 39278588 DOI: 10.1016/j.expneurol.2024.114950] [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: 07/01/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
Intracerebral hemorrhage (ICH) is a severe disease that often leads to disability and death. Neuroinflammatory response is a key causative factor of early secondary brain injury after ICH. AIM2 is a DNA-sensing protein that recognizes cytosolic double-stranded DNA and take a significant part in neuroinflammation. Mitochondrial DNA participates in the translation of proteins such as the respiratory chain in the mitochondria. Whether mtDNA is involved in forming AIM2 inflammasome after ICH remains unclear. We used mice to construct ICH model in vivo and we used BV2 microglial cells treated with oxyhemoglobin to simulate ICH in vitro. Following lentiviral transfection to overexpress AIM2 antagonist P202, a notable decrease was observed in the levels of AIM2 inflammasome-associated proteins, leading to a reduction in dead neurons surrounding the hematoma and an enhancement in long-term and short-term behavior of neurological deficits. We further explored whether mtDNA took part in the AIM2 activation after ICH. The cytosolic mtDNA level was down-regulated by the mitochondrial division protector Mdivi-1 and up-regulated by transfection of mtDNA into cytoplasm. We found the expression level of AIM2 inflammasome-related proteins and inflammatory cytokines release were regulated by the cytosolic mtDNA level. In conclusion, after ICH, the mtDNA content in the cytoplasm of microglia around the hematoma rises, causing AIM2 inflammation leading to neuronal apoptosis, which leads to neurological deficits in mice. On the other hand, P202 was able to block inflammatory vesicle activation and improve neurological function by preventing the interaction between AIM2 protein and mitochondrial DNA.
Collapse
Affiliation(s)
- Feng Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haojie Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xinyu Tao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Kun Dai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
38
|
Zhang Y, Zhang H, Jiang M, Cao X, Ge X, Song B, Lan J, Zhou W, Qi Z, Gu X, Liu J, Zheng Y, Li M, Ji X. Neuroprotection on ischemic brain injury by Mg 2+/H 2 released from endovascular Mg implant. Bioact Mater 2024; 42:124-139. [PMID: 39280580 PMCID: PMC11402188 DOI: 10.1016/j.bioactmat.2024.08.019] [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: 07/01/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Most acute ischemic stroke patients with large vessel occlusion require stent implantation for complete recanalization. Yet, due to ischemia-reperfusion injury, over half of these patients still experience poor prognoses. Thus, neuroprotective treatment is imperative to alleviate the ischemic brain injury, and a proof-of-concept study was conducted on "biodegradable neuroprotective stent". This concept is premised on the hypothesis that locally released Mg2+/H2 from Mg metal within the bloodstream could offer synergistic neuroprotection against reperfusion injury in distant cerebral ischemic tissues. Initially, the study evaluated pure Mg's neuroactive potential using oxygen-glucose deprivation/reoxygenation (OGD/R) injured neuron cells. Subsequently, a pure Mg wire was implanted into the common carotid artery of the transient middle cerebral artery occlusion (MCAO) rat model to simulate human brain ischemia/reperfusion injury. In vitro analyses revealed that pure Mg extract aided mouse hippocampal neuronal cell (HT-22) in defending against OGD/R injury. Additionally, the protective effects of the Mg wire on behavioral abnormalities, neural injury, blood-brain barrier disruption, and cerebral blood flow reduction in MCAO rats were verified. Conclusively, Mg-based biodegradable neuroprotective implants could serve as an effective local Mg2+/H2 delivery system for treating distant cerebral ischemic diseases.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Neurology and Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Hongkang Zhang
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Miaowen Jiang
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Xiaofeng Cao
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xiaoxiao Ge
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Baoying Song
- Department of Neurology and Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jing Lan
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Wenhao Zhou
- Biomaterials Research Center, Northwest Institute for Non-ferrous Metal Research, Xi'an, 710016, China
| | - Zhengfei Qi
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Xuenan Gu
- School of Bioengineering, Beihang University, Beijing, 100191, China
| | - Juzhe Liu
- The Key Laboratory of Resources and Environmental System Optimization, Ministry of Education, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 100096, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Ming Li
- Department of Neurology and Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xunming Ji
- Department of Neurology and Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
39
|
Luo Y, Yuan L, Liu Z, Dong W, Huang L, Liao A, Xie Y, Liu R, Lan W, Cai Y, Zhu W. Inhibition of PCSK9 Protects against Cerebral Ischemia‒Reperfusion Injury via Attenuating Microcirculatory Dysfunction. Neurochem Res 2024; 50:10. [PMID: 39548030 DOI: 10.1007/s11064-024-04272-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: 11/05/2023] [Revised: 08/16/2024] [Accepted: 09/18/2024] [Indexed: 11/17/2024]
Abstract
Proprotein convertase substilin/kexin type 9 (PCSK9), a pivotal protein regulating lipid metabolism, has been implicated in promoting microthrombotic formation and inflammatory cascades, thereby contributing to cardiovascular ischemia/reperfusion (I/R) injury. However, its involvement in cerebral I/R injury and its potential role in microcirculation protection remain unexplored. In this investigation, we utilized a middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model to simulate ischemic stroke. Different concentrations of evolocumab (1, 5, 10 mg/kg, i.v.), a PCSK9 inhibitor, were administered to assess its impact. Immunofluorescence staining was employed to analyze changes in the expression of occludin, claudin-5, thrombocyte, ICAM-1, VCAM-1, and CD45, providing insights into blood-brain barrier integrity, platelet adhesion, and immune cell infiltration. Moreover, the Morris water maze and elevated plus maze were utilized to evaluate neurological and behavioral functions in MCAO/R mice, shedding light on the effects of PCSK9 inhibition. Our findings revealed a surge in plasma PCSK9 levels post-MCAO/R, peaking at 24 h post-reperfusion. Evolocumab (10 mg/kg) treatment significantly mitigated brain infarction and neurological deficits, evidenced by enhanced locomotor function and reduced post-stroke anxiety. However, it did not ameliorate cognitive impairment following MCAO/R. Additionally, evolocumab administration led to diminished leakage of evans blue solution and upregulated expression of occludin and claudin-5. Thrombocyte, ICAM-1, VCAM-1, and CD45 levels were notably reduced in the penumbral area post-evolocumab treatment. These protective effects are speculated to be mediated through the inhibition of the ERK/NF-κB pathway. The PCSK9 inhibitor evolocumab holds promise as a therapeutic agent during the acute phase of stroke, exerting its beneficial effects by modulating the ERK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuanfei Luo
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Linying Yuan
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhihui Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weichen Dong
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Huang
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Anyu Liao
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yi Xie
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rui Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenya Lan
- Department of Cerebrovascular Disease Treatment Center, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yulong Cai
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Wusheng Zhu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
40
|
Zhang Y, Yang Q, Cheng H, Zhang Y, Xie Y, Zhang Q. Extracellular vesicles derived from endothelial progenitor cells modified by Houshiheisan promote angiogenesis and attenuate cerebral ischemic injury via miR-126/PIK3R2. Sci Rep 2024; 14:28166. [PMID: 39548169 PMCID: PMC11568282 DOI: 10.1038/s41598-024-78717-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Angiogenesis following cerebral ischemia is crucial for restoring blood supply to the ischemic region. Extracellular vesicles (EVs) derived from endothelial progenitor cells (EPCs) offer potential therapeutic benefits in the treatment of cerebral ischemia. Houshiheisan (HSHS) has been shown to improve clinical outcomes in ischemic stroke patients, reduce cerebral ischemic damage in rats, and protect endothelial cells. However, the potential effects of HSHS-modified EPC-derived EVs (EVsHSHS) for cerebral ischemia remain unexplored. This study investigated the impact of EVsHSHS on angiogenesis using rats with permanent middle cerebral artery occlusion (pMCAO) and brain microvascular endothelial cells (BMECs) subjected to oxygen-glucose deprivation (OGD). Results demonstrated that EVsHSHS promoted the proliferation, migration, and tube formation of BMECs in vitro. In vivo, high doses of EVsHSHS exhibited better performance than equivalent doses of unmodified EPC-derived EVs in reducing cerebral infarction volume, improving cortical blood perfusion, decreasing neurological deficit scores, and increasing cortical microvessel density at day 7 post-modeling. The pro-angiogenic effects of EVsHSHS following cerebral ischemia were associated with the regulation of miR-126 and the PIK3R2/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Yawen Zhang
- Department of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Qiuyue Yang
- Department of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Hongfa Cheng
- Department of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Yahui Xie
- Department of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Qiuxia Zhang
- Department of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
- Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China.
| |
Collapse
|
41
|
Tassinari ID, Zang J, Ribeiro NH, Martins BB, Tauffer JVM, Nunes RR, Sanches EF, Sizonenko S, Netto CA, Paz AH, de Fraga LS. Lactate administration causes long-term neuroprotective effects following neonatal hypoxia-ischemia. Exp Neurol 2024; 381:114929. [PMID: 39168170 DOI: 10.1016/j.expneurol.2024.114929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Neonatal hypoxia-ischemia (HI) is one of the main causes of mortality and long-term disabilities in newborns, and the only clinical approach to treat this condition is therapeutic hypothermia, which shows some limitations. Thus, putative neuroprotective agents have been tested in animal models of HI. Lactate is a preferential metabolic substrate of the neonatal brain and has already been shown to produce beneficial neuroprotective outcomes in neonatal animals exposed to HI. Here, we administered lactate as a treatment in neonatal rats previously exposed to HI and evaluated the impact of this treatment in adulthood. Seven-day-old (P7) male and female Wistar rats underwent permanent common right carotid occlusion combined with an exposition to a hypoxic atmosphere (8% oxygen) for 60 min. Animals were assigned to one of four experimental groups: HI, HI+LAC, SHAM, SHAM+LAC. Lactate was administered intraperitoneally 30 min and 2 h after hypoxia in HI+LAC and SHAM+LAC groups, whereas HI and SHAM groups received vehicle. Animals were tested in the behavioral tasks of negative geotaxis and righting reflex (P8), cylinder test (P24), and the modified neurological severity score was calculated (P25). Open field (OF), and novel object recognition (NOR) were evaluated in adulthood. Animals were killed at P60, and the brains were harvested and processed to evaluate the volume of brain injury. Our results showed that lactate administration reduced the volume of brain lesion and improved sensorimotor and cognitive behaviors in neonatal, juvenile, and adult life in HI animals from both sexes. Thus, lactate administration might be considered as a potential neuroprotective strategy for the treatment of neonatal HI, which is a prevalent disorder affecting newborns.
Collapse
Affiliation(s)
- Isadora D'Ávila Tassinari
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS 90035-903, Brazil
| | - Janaína Zang
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS 90035-903, Brazil
| | - Nícolas Heller Ribeiro
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil
| | - Bianca Büchele Martins
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil
| | - João Vitor Miotto Tauffer
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil
| | - Ricardo Ribeiro Nunes
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS 90035-903, Brazil
| | - Eduardo Farias Sanches
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Stéphane Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Departamento de Bioquímica, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil
| | - Ana Helena Paz
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Departamento de Ciências Morfológicas, Federal University of Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre 90050-170, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS 90035-903, Brazil
| | - Luciano Stürmer de Fraga
- Departamento de Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS 90035-903, Brazil.
| |
Collapse
|
42
|
Huang M, Cheng S, Li Z, Chen J, Wang C, Li J, Zheng H. Preconditioning Exercise Inhibits Neuron Ferroptosis and Ameliorates Brain Ischemia Damage by Skeletal Muscle-Derived Exosomes via Regulating miR-484/ACSL4 Axis. Antioxid Redox Signal 2024; 41:769-792. [PMID: 38545792 DOI: 10.1089/ars.2023.0492] [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] [Indexed: 04/25/2024]
Abstract
Aims: Although there is evidence that patients with stroke who exercise regularly before stroke have a better prognosis than those who do not exercise, the detailed mechanism remains unclear. Moreover, neuronal death plays a central role in neurological dysfunction caused by ischemic stroke. Thus, we investigated whether exercise could reduce stroke-induced neuronal death and its associated mediators in the current study. Results: Ferroptosis was the most dominant form of programmed cell death in neurons. Preconditioning exercise before stroke improved the neurological function and decreased the infarct area in rats with ischemic stroke. Preconditioning exercise attenuated stroke-induced ferroptosis by reducing lipid peroxidation (LPO) production, upregulating glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11), and downregulating acyl-CoA synthetase long-chain family member 4 (ACSL4). High-throughput sequencing and dual luciferase reporter assays revealed that exercise-induced exosomal miR-484 inhibits Acsl4 expression. Moreover, we showed that exercise-induced exosomal miR-484 is mainly derived from skeletal muscle, and the neuroprotective effect of preconditioning exercise is suppressed by inhibiting miR-484 production in skeletal muscle. Innovation: This study suggested that neuronal ferroptosis is the most dominant form of programmed cell death in a hypoxic environment. Moreover, we showed that the ferroptosis pathway is a potential therapeutic target in ischemic stroke and that preconditioning exercise could be an effective antioxidant intervention for cerebral ischemia. Conclusion: Our work revealed that preconditioning exercise before stroke exerts neuroprotective effects against brain ischemia by skeletal muscle-derived exosomal miR-484 via inhibiting ferroptosis. Antioxid. Redox Signal. 41, 769-792.
Collapse
Affiliation(s)
- Mudan Huang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shimei Cheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ziwen Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinshuo Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chuangjia Wang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jun Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
43
|
Hegde M, Singh AK, Kannan S, Kolkundkar U, Seetharam RN. Therapeutic Applications of Engineered Mesenchymal Stromal Cells for Enhanced Angiogenesis in Cardiac and Cerebral Ischemia. Stem Cell Rev Rep 2024; 20:2138-2154. [PMID: 39305405 PMCID: PMC11554727 DOI: 10.1007/s12015-024-10787-3] [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] [Accepted: 08/31/2024] [Indexed: 11/12/2024]
Abstract
Ischemic diseases are characterized by obstruction of blood flow to the respective organs, of which ischemia of the heart and brain are the most prominent manifestations with shared pathophysiological mechanisms and risk factors. While most revascularization therapies aim to restore blood flow, this can be challenging due to the limited therapeutic window available for treatment approaches. For a very long time, mesenchymal stromal cells have been used to treat cerebral and cardiac ischemia. However, their application is restricted either by inefficient mode of delivery or the low cell survival rates following implantation into the ischemic microenvironment. Nonetheless, several studies are currently focusing on using of mesenchymal stromal cells engineered to overexpress therapeutic genes as a cell-based gene therapy to restore angiogenesis. This review delves into the utilization of MSCs for angiogenesis and the applications of engineered MSCs for the treatment of cardiac and cerebral ischemia. Moreover, the safety issues related to the genetic modification of MSCs have also been discussed.
Collapse
Affiliation(s)
- Madhavi Hegde
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Suresh Kannan
- Stempeutics Research Pvt. Ltd., 3rd Floor, Manipal Hospitals Whitefield #143, EPIP Industrial Area, ITPL Main Road, Bangalore, 560 048, India
| | - Udaykumar Kolkundkar
- Stempeutics Research Pvt. Ltd., 3rd Floor, Manipal Hospitals Whitefield #143, EPIP Industrial Area, ITPL Main Road, Bangalore, 560 048, India
| | - Raviraja N Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India.
| |
Collapse
|
44
|
Vahidi S, Bigdeli MR, Shahsavarani H, Ahmadloo S, Roghani M. Neuroprotective Therapeutic Potential of microRNA-149-5p against Murine Ischemic Stroke. Mol Neurobiol 2024; 61:8886-8903. [PMID: 38573413 DOI: 10.1007/s12035-024-04159-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Ischemic stroke resulting from blockade of brain vessels lacks effective treatments, prompting exploration for potential therapies. Among promising candidates, microRNA-149 (miR-149) has been investigated for its role in alleviating oxidative stress, inflammation, and neurodegeneration associated with ischemic conditions. To evaluate its therapeutic effect, male Wistar rats were categorized into five groups, each consisting of 27 rats: sham, MCAO, lentiviral control, lentiviral miR-149, and miR149-5p mimic. Treatments were microinjected intracerebroventricularly (ICV) (right side), and ischemia was induced using middle cerebral artery occlusion (MCAO) procedure. Post-MCAO, neurological function, histopathological changes, blood-brain barrier (BBB) permeability, cerebral edema, and mRNA levels of Fas ligand (Faslg) and glutamate ionotropic NMDA receptor 1 (GRIN1) were assessed, alongside biochemical assays. MiR-149 administration improved neurological function, reduced brain damage, preserved BBB integrity, and attenuated cerebral edema. Upregulation of miR149-5p decreased Faslg and GRIN1 expression in ischemic brain regions. MiR-149 also reduced oxidative stress, enhanced antioxidant activity, decreased caspase-1 and - 3 activity, and modulated inflammatory factors in ischemic brain regions. Moreover, DNA fragmentation as an index of cell death decreased following miR-149 treatment. In conclusion, the study underscores miR-149 potential as a neuroprotective agent against ischemic stroke, showcasing its efficacy in modulating various mechanisms and supporting its candidacy as a promising therapeutic target for innovative strategies in stroke treatment.
Collapse
Affiliation(s)
- Samira Vahidi
- Department of Animal Science and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad-Reza Bigdeli
- Department of Animal Science and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
- Institute for Cognitive and Brain Science, Shahid Beheshti University, Tehran, Iran.
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Salma Ahmadloo
- Department of Animal Science and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
45
|
Zhu J, Xu N, Lin H, Deng L, Xie B, Jiang X, Liao R, Yang C. Remote ischemic preconditioning plays a neuroprotective role in cerebral ischemia-reperfusion mice by inhibiting mitophagy. Heliyon 2024; 10:e39076. [PMID: 39640619 PMCID: PMC11620096 DOI: 10.1016/j.heliyon.2024.e39076] [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: 03/14/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 12/07/2024] Open
Abstract
Remote ischemic preconditioning (RIPC) represents a clinically feasible method for safeguarding vital organs against ischemic injury. However, its specific role in cerebral ischemia-reperfusion (I/R) injury remains to be definitively elucidated. In this study, we investigated the neuroprotective effects of RIPC on mice at 7 days post-cerebral I/R and its involvement in mitophagy and mitochondrial dysfunction. Cerebral I/R led to impaired brain function, as well as structural and functional damage to mitochondria. Notably, RIPC treatment ameliorated the neurological dysfunction induced by cerebral I/R. Compared with the I/R group, the expression levels of NeuN, MBP, PDH, and Tom20 were significantly elevated in the RIPC + I/R group. Furthermore, mitochondria in the RIPC + I/R group exhibited more intact structure compared to those in the I/R group. In mice subjected to I/R injury, RIPC treatment markedly increased ATP content, ADP content, TAN level and glucose uptake while upregulating expression levels of Parkin, Pink1 and P62 proteins; it also reduced both the volume of ischemic foci and the number of mitochondrial autophagosomes along with decreasing LC3B II/I ratio. In conclusion, RIPC may exert a neuroprotective role by inhibiting excessive mitophagy during subacute stages following an ischemic stroke.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Na Xu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Heng Lin
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqian Jiang
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Runde Liao
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chaoxian Yang
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
46
|
Lei Q, Chen X, Xiong Y, Li S, Wang J, He H, Deng Y. Lysosomal Ca 2+ release-facilitated TFEB nuclear translocation alleviates ischemic brain injury by attenuating autophagic/lysosomal dysfunction in neurons. Sci Rep 2024; 14:24836. [PMID: 39438678 PMCID: PMC11496619 DOI: 10.1038/s41598-024-75802-6] [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: 05/27/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Neuronal death was frequently driven by autophagic/lysosomal dysfunction after ischemic stroke, whereas how to restore the impaired autophagic flux remained elusive. Autophagic/lysosomal signaling could be augmented after transcription factor EB (TFEB) nuclear translocation, which was facilitated by its dephosphorylation. A key TFEB dephosphorylase was calcineurin (CaN), whose activity was drastically regulated by cytosolic calcium ion concentration ([Ca2+]) controlled by lysosomal Ca2+ channel-like protein of TRPML1. Our research shows that ML-SA1, an agonist of the TRPML1 channel, significantly enhanced the lysosomal Ca2+ release and the CaN expression in penumbric neurons, subsequently promoted TFEB nuclear translocation, and greatly reversed autophagy/lysosome dysfunction. Moreover, ML-SA1 treatment significantly reduced neuronal loss, infarct size, and neurological deficits. By contrast, ML-SI3, an inhibitor of TRPML1, inhibited the lysosomal Ca2+ release conversely, aggravated the impairment of autophagic flux and consequentially exacerbated brain stroke lesion. These studies suggest that TRPML1 elevation alleviates ischemic brain injury by restoring autophagic/lysosomal dysfunction via Lysosomal Ca2+ release-facilitated TFEB nuclear translocation in neurons.
Collapse
Affiliation(s)
- Qian Lei
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xuemei Chen
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yajie Xiong
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China
| | - Shangdan Li
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jiaqian Wang
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China
| | - Hongyun He
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China.
- Anning First People's Hospital, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Yihao Deng
- School of Basic Medical Sciences, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
47
|
Liu G, Wang D, Jia J, Hao C, Ge Q, Xu L, Zhang C, Li X, Mi Y, Wang H, Miao L, Chen Y, Zhou J, Xu X, Liu Y. Neuroprotection of Human Umbilical Cord-Derived Mesenchymal Stem Cells (hUC-MSCs) in Alleviating Ischemic Stroke-Induced Brain Injury by Regulating Inflammation and Oxidative Stress. Neurochem Res 2024; 49:2871-2887. [PMID: 39026086 DOI: 10.1007/s11064-024-04212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Brain injury caused by stroke has a high rate of mortality and remains a major medical challenge worldwide. In recent years, there has been significant attention given to the use of human Umbilical cord-derived Mesenchymal Stem Cells (hUC-MSCs) for the treatment of stroke in different adult and neonate animal models of stroke. However, using hUC-MSCs by systemic administration to treat ischemic stroke has not been investigated sufficiently. In this study, we conducted various experiments to explore the neuroprotection of hUC-MSCs in rats. Our findings demonstrate that an intravenous injection of a high dose of hUC-MSCs at 2 × 10^7 cells/kg markedly ameliorated brain injury resulting from ischemic stroke. This improvement was observed one day after inducing transient middle cerebral artery occlusion (MCAO) and subsequent reperfusion in rats. Notably, the efficacy of this single administration of hUC-MSCs surpassed that of edaravone, even when the latter was used continuously over three days. Mechanistically, secretory factors derived from hUC-MSCs, such as HGF, BDNF, and TNFR1, ameliorated the levels of MDA and T-SOD to regulate oxidative stress. In particular, TNFR1 also improved the expression of NQO-1 and HO-1, important proteins associated with oxidative stress. More importantly, TNFR1 played a significant role in reducing inflammation by modulating IL-6 levels in the blood. Furthermore, TNFR1 was observed to influence the permeability of the blood-brain barrier (BBB) as demonstrated in the evan's blue experiment and protein expression of ZO-1. This study represented a breakthrough in traditional methods and provided a novel strategy for clinical medication and trials.
Collapse
Affiliation(s)
- Guangyang Liu
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Daohui Wang
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Jianru Jia
- Baoding People's Hospital, Baoding, China
| | - Chunhua Hao
- State Key Laboratory of Drug Delivery and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Qinggang Ge
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Liqiang Xu
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Chenliang Zhang
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Xin Li
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Yi Mi
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Herui Wang
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Li Miao
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Yaoyao Chen
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Jingwen Zhou
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Xiaodan Xu
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China
| | - Yongjun Liu
- Stem Cell Biology and Regenerative Medicine Institution, Beijing YiChuang Institute of Bio-Industry, Beijing, China.
| |
Collapse
|
48
|
Kim MJ, Lee D, Ryu JH, Lee SY, Choi BT, Yun YJ, Shin HK. Weisheng-tang protects against ischemic brain injury by modulating microglia activation through the P2Y12 receptor. Front Pharmacol 2024; 15:1347622. [PMID: 39295932 PMCID: PMC11408171 DOI: 10.3389/fphar.2024.1347622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Background: Stroke, a leading cause of death and disability, lacks effective treatments. Post-stroke secondary damage worsens the brain microenvironment, further exacerbating brain injury. Microglia's role in responding to stroke-induced damage in peri-infarct regions is crucial. In this study, we explored Weisheng-tang's potential to enhance ischemic outcomes by targeting microglia. Methods: We induced middle cerebral artery occlusion and reperfusion in mice, followed by behavioral assessments and infarct volume analyses after 48 h, and examined the changes in microglial morphology through skeleton analysis. Results: Weisheng-tang (300 mg/kg) significantly reduced infarction volume and alleviated neurological and motor deficits. The number of activated microglia was markedly increased within the peri-infarct territory, which was significantly reversed by Weisheng-tang. Microglial morphology analysis revealed that microglial processes were retracted owing to ischemic damage but were restored in Weisheng-tang-treated mice. This restoration was accompanied by the expression of the purinergic P2Y12 receptor (P2Y12R), a key regulator of microglial process extension. Weisheng-tang increased neuronal Kv2.1 clusters while suppressing juxtaneuronal microglial activation. The P2Y12R inhibitor-ticagrelor-eliminated the tissue and functional recovery that had been observed with Weisheng-tang after ischemic damage. Discussion: Weisheng-tang improved experimental stroke outcomes by modulating microglial morphology through P2Y12R, shedding light on its neuroprotective potential in ischemic stroke.
Collapse
Affiliation(s)
- Min Jae Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Dohee Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Ji Hye Ryu
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Seo-Yeon Lee
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Young Ju Yun
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| |
Collapse
|
49
|
Guo H, Li W, Yang Z, Xing X. E3 ubiquitin ligase MARCH1 reduces inflammation and pyroptosis in cerebral ischemia-reperfusion injury via PCSK9 downregulation. Mamm Genome 2024; 35:346-361. [PMID: 39115562 DOI: 10.1007/s00335-024-10055-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Pyroptosis has been regarded as caspase-1-mediated monocyte death that induces inflammation, showing a critical and detrimental role in the development of cerebral ischemia-reperfusion injury (IRI). MARCH1 is an E3 ubiquitin ligase that exerts potential anti-inflammatory functions. Therefore, the study probed into the significance of MARCH1 in inflammation and pyroptosis elicited by cerebral IRI. Middle cerebral artery occlusion/reperfusion (MCAO/R)-treated mice and oxygen glucose deprivation/reoxygenation (OGD/R)-treated hippocampal neurons were established to simulate cerebral IRI in vivo and in vitro. MARCH1 and PCSK9 expression was tested in MCAO/R-operated mice, and their interaction was identified by means of the cycloheximide assay and co-immunoprecipitation. The functional roles of MARCH1 and PCSK9 in cerebral IRI were subsequently determined by examining the neurological function, brain tissue changes, neuronal viability, inflammation, and pyroptosis through ectopic expression and knockdown experiments. PCSK9 expression was increased in the brain tissues of MCAO/R mice, while PCSK9 knockdown reduced brain damage and neurological deficits. Additionally, inflammation and pyroptosis were inhibited in OGD/R-exposed hippocampal neurons upon PCSK9 knockdown, accompanied by LDLR upregulation and NLRP3 inflammasome inactivation. Mechanistic experiments revealed that MARCH1 mediated ubiquitination and degradation of PCSK9, lowering PCSK9 protein expression. Furthermore, it was demonstrated that MARCH1 suppressed inflammation and pyroptosis after cerebral IRI by downregulating PCSK9 both in vivo and in vitro. Taken together, the present study demonstrate the protective effect of MARCH1 against cerebral IRI through PCSK9 downregulation, which might contribute to the discovery of new therapies for improving cerebral IRI.
Collapse
Affiliation(s)
- Hongmei Guo
- Department of Neurology, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, 1 Benxi Street, Qingshan District, Wuhan City, Hubei Province, 430080, China
| | - Wanli Li
- Department of Neurology, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, 1 Benxi Street, Qingshan District, Wuhan City, Hubei Province, 430080, China
| | - Zhigang Yang
- Department of Neurology, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, 1 Benxi Street, Qingshan District, Wuhan City, Hubei Province, 430080, China
| | - Xiaobin Xing
- Department of Neurology, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, 1 Benxi Street, Qingshan District, Wuhan City, Hubei Province, 430080, China.
| |
Collapse
|
50
|
Yang X, Mu Y, Feng Y, Li M, Hu H, Zhang X, Zuo Z, Wu R, Xu J, Zheng F, He X, Hu X, Zhang L. Physical exercise-induced circAnks1b upregulation promotes protective endoplasmic reticulum stress and suppresses apoptosis via miR-130b-5p/Pak2 signaling in an ischemic stroke model. CNS Neurosci Ther 2024; 30:e70055. [PMID: 39328024 PMCID: PMC11427801 DOI: 10.1111/cns.70055] [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: 03/05/2024] [Revised: 08/02/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
AIMS Physical exercise (PE) can accelerate post-stroke recovery. This study investigated contributions of circRNAs to PE-induced improvements in post-stroke neurological function. METHODS Rats subjected to transient middle cerebral artery occlusion were left sedentary or provided running-wheel access for 4 weeks during recovery. CircRNAs from peri-infarct cortex were identified by high-throughput sequencing, and interactions with miRNAs by immunoprecipitation, fluorescence in suit hybridization, and dual-luciferase reporter assays. In vivo circRNA knockdown was achieved using shRNA-AAVs and in vitro overexpression by plasmid transfection. Transmission electron microscopy, western blotting, and TUNEL assays were conducted to explore circRNA contributions to endoplasmic reticulum (ER) stress and neuronal apoptosis. CircRNA levels were measured in plasma from stroke patients by qRT-PCR and associations with neurological scores assessed by Pearson's correlation analysis. RESULTS PE upregulated circAnks1b, reduced infarct volume, and mitigated neurological dysfunction, while circAnks1b knockdown exacerbated neurological dysfunction and increased infarct size despite PE. CircAnks1b sponged miR-130b-5p, thereby disinhibiting Pak2 expression. Conversely, Pak2 downregulation disrupted PE-mediated protective ER stress, leading to reduced IRE1/XBP1 and heightened apoptosis. Plasma circAnks1b was higher in stroke patients receiving PE than sedentary patients and correlated negatively with neurological scores. CONCLUSIONS CircAnks1b upregulation may be an effective therapeutic strategy for post-stroke recovery.
Collapse
Affiliation(s)
- Xiaofeng Yang
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Yating Mu
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Yifeng Feng
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Mingyue Li
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Haojie Hu
- Department of PsychologyNew York UniversityNew YorkNew YorkUSA
| | - Xiaoya Zhang
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zejie Zuo
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Rui Wu
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Jinghui Xu
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Fang Zheng
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Xiaofei He
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Xiquan Hu
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Liying Zhang
- Department of Rehabilitation MedicineThe Third Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|