1
|
Holm H, Jujic A, Nilsson PM, Magnusson M, Malan L. The prognostic significance of stress-phenotyping for stroke incidence: the Malmö Diet and Cancer Study. Stress 2025; 28:2443980. [PMID: 39731532 DOI: 10.1080/10253890.2024.2443980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Self-reported mental stress is not consistently recognized as a risk factor for stroke. This prompted development of a novel algorithm for stress-phenotype indices to quantify chronic stress prevalence in relation to a modified stroke risk score in a South African cohort. The algorithm is based on biomarkers adrenocorticotrophic hormone, high-density lipoprotein cholesterol, high-sensitive cardiac-troponin-T, and diastolic blood pressure which exemplifies the stress-ischemic-phenotype index. Further modification of the stroke risk score to accommodate alcohol misuse established the stress-diabetes-phenotype index. Whether positive stress-phenotype individuals will demonstrate a higher incidence of stroke in an independent Swedish cohort was unknown and investigated. METHODS Stress-phenotyping was done at baseline for 50 participants with incident stroke and 100 age-, and sex matched controls (aged 76 ± 5 years) from 2,924 individuals in southern Sweden. The mean time from inclusion to first stroke event was 5 ± 3 years. Stress-phenotyping comparisons and stroke incidence risk were determined. RESULTS A positive stress-ischemic-phenotype reflected higher incident stroke (72% vs. 28%, p = 0.019) and mortality rates (41% vs. 23%, p = 0.019). Whereas a positive stress-diabetes-phenotype reflected a higher incident stroke rate (80% vs. 20%, p = 0.008) but similar mortality rate (38% vs. 25%, p = 0.146). Both the positive stress-ischemic (OR: 2.9, 95% CI: 1.3-6.5, p = 0.011) and stress-diabetes-phenotypes (OR: 3.7, 95% CI: 1.5-8.9, p = 0.004) showed large effect size associations with incident stroke independent of cardiovascular risk confounders. CONCLUSION Positive stress-phenotype indices demonstrated a higher incidence of stroke. Ultimately the Malan stress-phenotype algorithms developed in South Africa could confirm incident stroke in an independent Swedish cohort. Stress-phenotyping could thus be useful in clinical routine practice in order to detect individuals at higher stroke risk.
Collapse
Affiliation(s)
- H Holm
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - A Jujic
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - P M Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - M Magnusson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - L Malan
- Technology Transfer and Innovation-Support Office, North-West University, Potchefstroom, South Africa
| |
Collapse
|
2
|
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
|
3
|
Zheng J, Li Y, Zhang T, Fu Y, Long P, Gao X, Wang Z, Guan Z, Qi X, Hong W, Xiao Y. Endoplasmic reticulum stress and autophagy in cerebral ischemia/reperfusion injury: PERK as a potential target for intervention. Neural Regen Res 2025; 20:1455-1466. [PMID: 39075912 PMCID: PMC11624856 DOI: 10.4103/nrr.nrr-d-23-00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/21/2023] [Accepted: 11/21/2023] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00028/figure1/v/2024-07-28T173839Z/r/image-tiff Several studies have shown that activation of unfolded protein response and endoplasmic reticulum (ER) stress plays a crucial role in severe cerebral ischemia/reperfusion injury. Autophagy occurs within hours after cerebral ischemia, but the relationship between ER stress and autophagy remains unclear. In this study, we established experimental models using oxygen-glucose deprivation/reoxygenation in PC12 cells and primary neurons to simulate cerebral ischemia/reperfusion injury. We found that prolongation of oxygen-glucose deprivation activated the ER stress pathway protein kinase-like endoplasmic reticulum kinase (PERK)/eukaryotic translation initiation factor 2 subunit alpha (eIF2α)-activating transcription factor 4 (ATF4)-C/EBP homologous protein (CHOP), increased neuronal apoptosis, and induced autophagy. Furthermore, inhibition of ER stress using inhibitors or by siRNA knockdown of the PERK gene significantly attenuated excessive autophagy and neuronal apoptosis, indicating an interaction between autophagy and ER stress and suggesting PERK as an essential target for regulating autophagy. Blocking autophagy with chloroquine exacerbated ER stress-induced apoptosis, indicating that normal levels of autophagy play a protective role in neuronal injury following cerebral ischemia/reperfusion injury. Findings from this study indicate that cerebral ischemia/reperfusion injury can trigger neuronal ER stress and promote autophagy, and suggest that PERK is a possible target for inhibiting excessive autophagy in cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ju Zheng
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
- Guizhou Center for Disease Control and Prevention, Guiyang, Guizhou Province, China
| | - Yixin Li
- Department of Histology and Embryology, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Ting Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yanlin Fu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Peiyan Long
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xiao Gao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Zhengwei Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Zhizhong Guan
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou Province, China
| | - Wei Hong
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou Province, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou Province, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou Province, China
| |
Collapse
|
4
|
Ju J, Cheng C, Cui L, Hong B, Zhan Q, Wang Q, Cui X, Su D, Huang Y, Kang C. EPIC-1042 alleviates cerebral ischemic/reperfusion injury through TAX1BP1-induced mitophagy. Free Radic Biol Med 2025; 232:367-381. [PMID: 40107570 DOI: 10.1016/j.freeradbiomed.2025.03.023] [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: 01/09/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Post ischemia-reperfusion (I/R) injury, an upregulation in Polymerase I and transcript release factor (PTRF) expression is observed. PTRF is implicated in the regulation of various cellular processes within neuronal cells, thereby exacerbating the deleterious effects of I/R injury. EPIC-1042 is a small molecule pharmacological agent that exhibits specificity in binding to PTRF. Therefore, this study aimed to explore whether EPIC-1042 could be used as a treatment for I/R injury. To achieve this goal, we observed brain injury in mice following EPIC-1042 pre-administration, and then transitioned to therapeutic administration. After observing the pre-protective and therapeutic effects of the drug, proteomic analysis revealed that the expression of TAX1BP1 continued to decline in a time-dependent manner, while EPIC-1042 was able to inhibit this decline. However, the function of TAX1BP1 in ischemic stroke is not yet fully understood. Subsequent experiments confirmed that the addition of EPIC-1042 resulted in an enhancement of mitophagy. Silencing the expression of TAX1BP1 abrogated the drug's effects, indicating that EPIC-1042 exerts a protective function by promoting mitophagy via TAX1BP1 mediation. We further investigated the synergistic effects of EPIC-1042 and edaravone by administering the two drugs in combination, observing an enhanced therapeutic efficacy compared to the administration of each drug alone. Subsequently, we optimized the administration protocol for the two drugs by utilizing liposome encapsulation for both drugs. This approach enabled us to achieve significant therapeutic outcomes while reducing both the dosage and frequency of administration, thereby demonstrating the potential for clinical translation of EPIC-1042.
Collapse
Affiliation(s)
- Jiasheng Ju
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Chunchao Cheng
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Longtao Cui
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Biao Hong
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Qi Zhan
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Qixue Wang
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Xiaoteng Cui
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Dongyuan Su
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Yanping Huang
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Chunsheng Kang
- General Hospital of Tianjin Medical University, Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| |
Collapse
|
5
|
Tsai TH, Chang YT, Cheng YC. Association of cataract surgery with stroke among older adults in the United States. Eye (Lond) 2025; 39:1400-1405. [PMID: 39922969 PMCID: PMC12044012 DOI: 10.1038/s41433-025-03662-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: 05/31/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cataract surgery, one of the most frequent conducted surgeries around the world, is associated with cardiovascular diseases. We aim to determine the association of cataract surgery and the risk of stroke. METHODS Adults aged over 65 years old in the National Health and Aging Trends Study were followed 7 years annually. There were 6700 stroke-free participants included at baseline survey. These participants were divided into two groups based on past history of cataract surgery. Demographics and multiple comorbidities were compared between the two groups. We identified newly developed cases of stroke over a 7-year period and performed survival analysis. Cox regression was further performed to yield adjusted hazard ratios. RESULTS Among 6700 elderly participants, 2803 of them had a history of cataract surgery while 3897 of them had not. Over the 7-year follow-up period, the cumulative stroke-free survival rate among the cataract surgery group and the control group were 84.4% versus 88.6% (p < 0.0001, log-rank test). Compared with the control group, elderly with a history of cataract surgery had a higher risk of developing stroke (adjusted HR 1.36, 95% CI 1.03 to 1.79, p = 0.026) after adjusting for multiple covariates. Other significant predictors included age ≥80 years old, having comorbidities with heart disease, lung disease, and dementia. Contrarily, protective factors for further stroke development included higher education and more frequent outdoor activities. CONCLUSIONS Our findings suggest that patients with a history of cataract surgery had a 1.36-fold increased risk of future stroke development.
Collapse
Affiliation(s)
- Tsung-Hsien Tsai
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yuan-Ting Chang
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Chen Cheng
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan.
- Department of Neurology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, 24352, Taiwan.
| |
Collapse
|
6
|
Xiong Z, Ou Y, Chen R, Zhou M, Wang Z, Wu G, Che M, Li K, Gong H, Wang Y, Ling X, Wang H, Wang X, Song Q, Qi S, Feng Z, Peng J. Tanycyte proliferation and migration through the sonic hedgehog pathway restores hypothalamic function after ischemic injury. Free Radic Biol Med 2025; 232:437-449. [PMID: 40113020 DOI: 10.1016/j.freeradbiomed.2025.03.026] [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: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Tanycytes, a distinct type of glial cell within the hypothalamus, will be investigated in this study to elucidate the intrinsic mechanisms by which they facilitate the restoration of hypothalamic function. We injected endothelin 1 (ET-1) into the third ventricle to establish an ischemic hypothalamic injury model. Nestin CreERT2 and Rosa26R-CAG:tdTomato mice were crossbred, and viral tracing was used to label and track tanycytes. Functional changes in these cells were observed with calcium imaging. Alterations in tanycytes were assessed with single-cell and transcriptomic sequencing analyses. The involvement of specific pathways was confirmed via intraperitoneal injection of N-acetyl cysteine (NAC) and cycloheximide. Following ischemic injury to the hypothalamus in mice, acute weight loss and impaired activity of Agrp neurons were observed, both of which recovered within 7 days. The fate of tanycytes was traced in Nestin-CreERT2: Rosa26R-CAG:Tdtomato mice to confirm their proliferation and migration after hypothalamic injury. Calcium imaging indicated that these proliferating and migrating cells participated in signal transduction, thereby reconstructing the regulatory network of tanycytes. The analysis of single-cell data on postnatal days 8 and 45 identified CDK1 as a marker of proliferative tanycytes. The roles of ROS and the Shh pathway in the proliferation and migration of tanycytes were validated via the intraperitoneal injection of NAC and cycloheximide inhibitors. After inducing ischemic injury to the arcuate nucleus of the hypothalamus, Agrp neuronal activity declined, accompanied by ROS fluctuations within tanycytes. Activation of the Shh pathway prompts the transition of tanycytes from a quiescent state to a proliferative state, thereby leading to their migration to the arcuate nucleus. This process re-establishes the regulatory network of tanycytes and restores metabolic balance. This finding may provide an important target for promoting the recovery of hypothalamic function.
Collapse
Affiliation(s)
- Zhiwei Xiong
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yichao Ou
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Rongjun Chen
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Mingfeng Zhou
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zijing Wang
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Guangsen Wu
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Mengjie Che
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Kai Li
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Haodong Gong
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yihan Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Xufan Ling
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Hai Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Xingqin Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Qiancheng Song
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China.
| | - Zhanpeng Feng
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China.
| | - Junxiang Peng
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Liu J, Qing T, He M, Xu L, Wu Z, Huang M, Liu Z, Zhang Y, Li Z, Yang W, Liu J, Li J. Transcriptomics, single-cell sequencing and spatial sequencing-based studies of cerebral ischemia. Eur J Med Res 2025; 30:326. [PMID: 40275374 PMCID: PMC12020253 DOI: 10.1186/s40001-025-02596-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
With high disability and mortality rate as well as highly complex pathogenesis, cerebral ischemia is highly morbid, prone to recurrence. To comprehensively understand the pathophysiological process of cerebral ischemia and to find new therapeutic strategies, a new approach to cerebral ischemia transcriptomics has emerged in recent years. By integrating data from multiple levels of transcriptomics, such as transcriptomics, single-cell transcriptomics, and spatial transcriptomics, this new approach can provide powerful help in revealing the molecular mechanisms of cerebral ischemia occurrence and development. Key findings highlight the critical roles of inflammation, blood-brain barrier dysfunction, and mitochondrial dysregulation in cerebral ischemia, offering potential biomarkers and therapeutic targets for early diagnosis and personalized treatment. A review of the research progress of cerebral ischemic injury mechanism under the analysis of the comprehensive transcriptomics research method was presented in this article, aiming to study the potential mechanism to provide new, innovative therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Jiaming Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Tao Qing
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Mei He
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory of Birth Defects Research and Prevention, Changsha, Hunan, China
| | - Liu Xu
- International Education School, Hunan University of Medicine, Huaihua, Hunan, China
| | - Zhuxiang Wu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Meiting Huang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Zheyu Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Ye Zhang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Zisheng Li
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Wenhui Yang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Junbo Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Jie Li
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China.
- Huaihua Key Laboratory of Ion Channels and Complex Diseases, Huaihua, Hunan, China.
| |
Collapse
|
8
|
An H, Shao C, He Y, Zhou H, Wang T, Xu G, Yang J, Wan H. Calycosin Inhibit PANoptosis and Alleviate Brain Damage: A Bioinformatics and Experimental Verification Approach. ACS Chem Neurosci 2025; 16:1550-1564. [PMID: 40156525 DOI: 10.1021/acschemneuro.5c00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
Abstract
PANoptosis is a newly identified form of cell death that encompasses pyroptosis, apoptosis, and necroptosis. Numerous studies have highlighted the significance of PANoptosis in brain ischemia-reperfusion (I/R) injury. Calycosin, a natural product with diverse biological activities, has demonstrated a significant reduction in neuronal death caused by ischemic brain injury by modulating multiple cell death pathways. In order to investigate the potential mechanisms underlying the neuroprotective role of calycosin in alleviating PANoptosis-induced damage in ischemic stroke therapy, we used mouse hippocampal neuronal cell line HT22 to stimulate ischemia in vitro through Oxygen and Glucose Deprivation/Reperfusion (OGD/R) and established molecular docking to assess the binding affinity of Calycosin with key targets and molecular dynamics simulations (MDS) to study the stability of the ligand-protein complex. The results demonstrate that Calycosin could improve the cell growth of HT22, leading to enhanced cell viability, reduced lactate dehydrogenase leakage, and decreased cell apoptosis after OGD/R. It also regulated the expression of PANoptosis-related genes such as NLRP3, GSDMD, MLKL, and RIPK1 and increased the Bcl-2/Bax ratio, effectively reducing cellular damage and providing protection. Molecular docking and MDS simulations demonstrated strong binding activity and stability between Calycosin and PANoptosis-related targets. Furthermore, Calycosin successfully passed the drug similarity (DS) evaluation and exhibited favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties and biological activity. In conclusion, Calycosin could alleviate ischemic stroke by inhibiting PANoptosis, reducing neuronal inflammation and apoptosis, and improving damage caused by the OGD/R. Thus, it could serve as a potential therapy for ischemic stroke.
Collapse
Affiliation(s)
- Huiyan An
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Chongyu Shao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Huifen Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Ting Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Guanfeng Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
- School of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| |
Collapse
|
9
|
Gao N, Huang Z, Xie J, Gao S, Wang B, Feng H, Bao C, Tian H, Liu X. Cryptotanshinone alleviates cerebral ischemia reperfusion injury by regulating ferroptosis through the PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119800. [PMID: 40222690 DOI: 10.1016/j.jep.2025.119800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/28/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Cryptotanshinone (CT) is a kind of Chinese medicine extracted from salvia miltiorrhiza, which has various pharmacological activities and is widely used in the treatment of diseases. AIM OF THE STUDY The objective is to delve into the mechanism by which cryptotanshinone (CT) exerts its effects on rats with the middle cerebral artery occlusion/reperfusion (MCAO/R) model. Additionally, it aims to further assess the interplay between inflammation and oxidative stress, along with the underlying mechanism of CT's anti-ferroptosis function. MATERIALS AND METHODS We constructed the middle cerebral artery occlusion/reperfusion (MCAO/R) model in rats. The effects of cryptotanshinone (CT) were evaluated using 2,3,5 - triphenyltetrazolium chloride (TTC) staining, behavioral assays, immunofluorescence, hematoxylin - eosin (HE) staining, and Nissl staining. Additionally, in vitro, cell viability was assessed by the Cell Counting Kit - 8 (CCK - 8) assay following experimental dosing. Oxygen - glucose deprivation/oxidation (OGD/R) models were established in PC12 and BV2 cells. Flow cytometry was employed to detect cellular reactive oxygen species (ROS) expression. The activities of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-px), and Mitochondrial Membrane Potential Assay Kit with JC-1(JC-1) were measured using biochemical methods. Inflammatory factor levels were determined using enzyme-linked immunosorbent assay (ELISA) kits. Immunoblotting was used to detect the levels of rat phosphatidylinositol 3 - kinase (PI3K), phosphorylated-PI3K (P-PI3K), protein kinase B (AKT), phosphorylated - AKT (P-AKT), nuclear factor erythroid 2 - related factor 2 (Nrf2), solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4). RESULTS In rats with the MCAO/R model, CT demonstrated the ability to decrease ROS levels, enhance the activity of glutathione (GSH), mitigate inflammation, augment the activity of glutathione peroxidase 4 (GPX4), inhibit ferroptosis, safeguard neurons, and facilitate the restoration of nerve function. Results from network pharmacology indicated that the action of CT might be mediated via the PI3K/Akt signaling pathway. Simultaneously, in-vivo investigations revealed that CT curbs ferroptosis through the PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathways. CONCLUSION CT can inhibit ferroptosis by inhibiting the vicious cycle between oxidative stress and inflammation, protect neurons and promote motor function recovery.
Collapse
Affiliation(s)
- Nana Gao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Zongyu Huang
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Jianjie Xie
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Shuang Gao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Biaobiao Wang
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Huicong Feng
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Cuifen Bao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China.
| | - He Tian
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China; Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China.
| | - Xia Liu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China.
| |
Collapse
|
10
|
Fu X, Li J, Yang S, Jing J, Zheng Q, Zhang T, Xu Z. Blood-brain barrier repair: potential and challenges of stem cells and exosomes in stroke treatment. Front Cell Neurosci 2025; 19:1536028. [PMID: 40260076 PMCID: PMC12009835 DOI: 10.3389/fncel.2025.1536028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
Stroke is characterized with high morbidity, mortality and disability all over the world, and one of its core pathologies is blood-brain barrier (BBB) dysfunction. BBB plays a crucial physiological role in protecting brain tissues and maintaining homeostasis in central nervous system (CNS). BBB dysfunction serves as a key factor in the development of cerebral edema, inflammation, and further neurological damage in stroke patients. Currently, stem cells and their derived exosomes have shown remarkable potential in repairing the damaged BBB and improving neurological function after stroke. Stem cells repair the integrity of BBB through anti-inflammatory, antioxidant, angiogenesis and regulation of intercellular signaling mechanisms, while stem cell-derived exosomes, as natural nanocarriers, further enhance the therapeutic effect by carrying active substances such as proteins, RNAs and miRNAs. This review will present the latest research advances in stem cells and their exosomes in stroke treatment, as well as the challenges of cell source, transplantation timing, dosage, and route of administration in clinical application, aiming to discuss their mechanisms of repairing BBB integrity and potential for clinical application, and proposes future research directions. Stem cells and exosomes are expected to provide new strategies for early diagnosis and precise treatment of stroke, and promote breakthroughs in the field of stroke.
Collapse
Affiliation(s)
- Xiaochen Fu
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Jia Li
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Shoujun Yang
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiapeng Jing
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Qinzhi Zheng
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Ting Zhang
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Zhuo Xu
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
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] [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
|
12
|
Zhu L, Zhong W, Meng X, Lv X, Deng T, Mei Z, Liu X, Meng F, Tian Y, Hu L, Xiang H, Chen Y, Li Y. Clusterzyme-Enabled Oxidative Stress Alleviation and Microglial Polarization Modulation for Efficient Ischemic Stroke Treatment. Adv Healthc Mater 2025; 14:e2404268. [PMID: 39998259 DOI: 10.1002/adhm.202404268] [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: 10/31/2024] [Revised: 01/27/2025] [Indexed: 02/26/2025]
Abstract
Ischemic stroke (IS) presents a significant challenge to global health, as conventional reperfusion strategies aimed at restoring cerebral circulation paradoxically exacerbate neurological damage. This injury primarily results from the excessive production of reactive oxygen species (ROS) and the initiation of a widespread neuroinflammatory response. In this study, mercaptosuccinic acid (MSA)-coated bimetallic clusterzymes, containing an optimized ratio of Au7Ag1 nanoclusters (NCs), are developed for the targeted treatment of IS reperfusion injury. The ultrafine particle size of bimetallic nanoclusters facilitates the penetration across the blood-brain barrier (BBB) and enhances catalytic capacity and enzymatic activity through synergistic effects. Comprehensive in vitro and in vivo studies demonstrate that Au7Ag1 NCs provide neuroprotection by efficiently scavenging ROS and modulating microglial polarization, alleviating oxidative stress-induced injury. Furthermore, Au7Ag1 NCs play a crucial role in reducing brain tissue damage following reperfusion and promoting long-term neurological function recovery. Notably, RNA sequencing reveals that Au7Ag1 NCs impact key molecular pathways linked to apoptosis and inflammation. In summary, this study demonstrates the potential of Au7Ag1 NCs as a novel therapeutic approach for IS reperfusion injury and highlights a promising pathway for nanomedicine-based interventions targeting ischemic cerebral disorders.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weijie Zhong
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuchen Meng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xin Lv
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tanjun Deng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zixian Mei
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xinru Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Fanying Meng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Yayuan Tian
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lan Hu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu, 215200, China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yi Li
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
13
|
Mojaver A, Khazaei M, Ahmadpanah M, Zarei M, Soleimani Asl S, Habibi P, Shahidi S. Dietary intake of coenzyme Q10 reduces oxidative stress in patients with acute ischemic stroke: a double-blind, randomized placebo-controlled study. Neurol Res 2025; 47:232-241. [PMID: 39999976 DOI: 10.1080/01616412.2025.2470712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 02/15/2025] [Indexed: 02/27/2025]
Abstract
OBJECTIVES Ischemic stroke is one of the most common neurological disorders. Oxidative stress, inflammation, and the reduction of Brain-Derived Neurotrophic Factor (BDNF) are implicated in cell death during ischemic stroke. Several studies suggest that Coenzyme Q10 (CoQ10) has antioxidant, anti-inflammatory, neuroprotective properties and can increase BDNF levels. This study investigated the effects of oral CoQ10 supplementation on oxidative stress biomarkers Total Antioxidant Capacity (TAC), Superoxide Dismutase (SOD), Malondialdehyde (MDA), Total Thiol Groups (TTG) - as well as serum levels of Interleukin-6 (IL-6) and BDNF in ischemic stroke patients. METHODS Fifty patients hospitalized for acute ischemic stroke were randomly divided into two groups: placebo (n = 25) and CoQ10 (600 mg/day) supplementation (n = 25). The intervention began 24 hours after stroke onset and continued for 30 days. RESULTS Significant reductions in serum MDA and IL-6 levels, alongside increased SOD and BDNF levels, were observed in the CoQ10 group. No significant differences were found in TAC or TTG levels between the groups. CONCLUSIONS A 30-day regimen of CoQ10 (600 mg/day) resulted in reduced oxidative stress and inflammation, alongside increased BDNF, suggesting potential neuroprotective benefits for post-stroke rehabilitation. CoQ10 May be considered a therapeutic option for enhancing neuroprotection and rehabilitation in stroke patients.
Collapse
Affiliation(s)
- Ali Mojaver
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mojtaba Khazaei
- Department of Neurology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Ahmadpanah
- Department of Clinical Psychology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Zarei
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Soleimani Asl
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parisa Habibi
- Department Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Institute of Neuroscience and Mental Health, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
14
|
Shadman J, Haghi-Aminjan H, Alipour MR, Panahpour H. The Neuroprotective Mechanisms of Kaempferol in Experimental Ischemic Stroke: A Preclinical Systematic Review. Mol Neurobiol 2025:10.1007/s12035-025-04848-y. [PMID: 40120044 DOI: 10.1007/s12035-025-04848-y] [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: 07/23/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Ischemic stroke represents a critical global health challenge, resulting in significant mortality and disability worldwide, yet there are limited effective treatment options currently available. While the intricate molecular pathways underlying the onset and progression of ischemic stroke are multifaceted, relying on a single therapeutic approach is unlikely to yield effective treatment for this complex disease. Therefore, it is crucial to explore efficient strategies that employ multifaceted targeting and address the multifarious pathological processes to overcome the challenges associated with ischemic brain injury. In recent times, natural plant-derived compounds have garnered significant interest as promising neuroprotective agents for the management of neurological conditions, including ischemic stroke. This study investigates the possible neuroprotective properties of kaempferol, a naturally occurring flavonoid compound, in mitigating the detrimental consequences of cerebral ischemic events. The findings from the reviewed preclinical studies suggest that kaempferol exhibits significant neuroprotective potential as a multifaceted therapeutic agent for ischemic stroke. Its efficacy stems from a combination of antioxidant, anti-inflammatory, and anti-apoptotic properties, which collectively mitigate ischemic stroke-induced brain injury. While these results are promising, clinical studies are essential to validate kaempferol's therapeutic viability for ischemic stroke patients.
Collapse
Affiliation(s)
- Javad Shadman
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | | | - Hamdollah Panahpour
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
15
|
Sharma D, Singh V, Kumar A, Singh TG. Genistein: A promising ally in combating neurodegenerative disorders. Eur J Pharmacol 2025; 991:177273. [PMID: 39828018 DOI: 10.1016/j.ejphar.2025.177273] [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: 06/27/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Neurodegenerative disorders arise when nerve cells in the brain or peripheral nervous system gradually lose functions and eventually die. Although certain therapies may alleviate some of the physical and mental symptoms associated with neurodegenerative disorders, hence slowing their progression, but no sure-shot treatment is currently available. It was shown that the rise in life expectancy and the number of elderly people in the community led to an increasing trend in the incidence and prevalence of neurodegenerative disease. Phytomolecules are demonstrating their effectiveness in combating, regression, and delaying various diseases. Genistein is one of soy isoflavone with antioxidant, anti-inflammatory, and estrogenic effects. Researchers demonstrated that Genistein treatment significantly reduced hyperglycemia, improved cognitive performance by modulating acetylcholinesterase activity and oxidative stress, and alleviated neuroinflammatory conditions in mice. This paper evaluates (in vivo and in vitro) various molecular targets of isoflavones and their ability to effectively counter several neurodegenerative disorders such as Parkinson's, Alzheimer's, and Huntington's diseases and amyotrophic lateral sclerosis. In this review, we aim to provide an overview of the role that genistein plays in delaying the development of neurodegenerative disorders.
Collapse
Affiliation(s)
- Diksha Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Varinder Singh
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India.
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | | |
Collapse
|
16
|
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] [MESH Headings] [Grants] [Track Full Text] [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
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
|
17
|
Huang Y, Yang J, Lu T, Shao C, Wan H. Puerarin Alleviates Cerebral Ischemia-Reperfusion Injury by Inhibiting Ferroptosis Through SLC7A11/GPX4/ACSL4 Axis and Alleviate Pyroptosis Through Caspase-1/GSDMD Axis. Mol Neurobiol 2025:10.1007/s12035-025-04798-5. [PMID: 40056342 DOI: 10.1007/s12035-025-04798-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
Cerebral ischemia-reperfusion (CIRI) represents a complex disease entity that encompasses multiple pathways. The occurrence of CIRI induces cerebral infarction, accompanied by brain tissue necrosis and focal neuronal impairment. Previous studies have demonstrated that ferroptosis, a specific cell death pathway implicated in CIRI, plays a crucial role in mediating the pathophysiological process of this condition. Puerarin, is known to possess vasodilatory, antioxidant, and neuroprotective properties. However, its precise role in ferroptosis as well as the underlying mechanisms remains elusive. In this study, we delved into the neuroprotective mechanisms of puerarin using both the rat middle cerebral artery occlusion (MCAO) model and the HT22 cell model of oxygen-glucose deprivation/reperfusion (OGD/R). In the MCAO model, puerarin was found to exhibit an inhibitory effect on ACSL4, which was consistent with that of rosiglitazone. Simultaneously, it was capable of counteracting the inhibition of GPX4 by RSL3. These findings suggest that puerarin modulates GPX4 and ACSL4, thereby exerting an inhibitory effect on ferroptosis. The ferroptosis-protective effect of puerarin was further corroborated in the OGD/R through a positive control experiment with ferrostatin-1, a lipid peroxidation inhibitor. Furthermore, we also recognized the importance of other cell death modalities, such as pyroptosis. Consequently, we verified the neuroprotective effect of puerarin by examining the influence of caspase-1 and GSDMD in HT22. Mechanistically, puerarin alleviates CIRI by respectively inhibiting ferroptosis through the SLC7A11/GPX4/ACSL4 axis and pyroptosis through the caspase-1/GSDMD axis. This research provides novel insights into the targeting and therapeutic potential of puerarin for the treatment of CIRI.
Collapse
Affiliation(s)
- Ying Huang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jiehong Yang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou, 310053, Zhejiang, China
| | - Ting Lu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Chongyu Shao
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou, 310053, Zhejiang, China.
| | - Haitong Wan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
18
|
Yue Q, Cao Z, Zhang T, Yin N, Liu L. Large Fibrous Connective Tissue Reduces Oxidative Stress to Form a Living Cell Scaffold in Adipose Grafts. Antioxidants (Basel) 2025; 14:270. [PMID: 40227213 PMCID: PMC11939587 DOI: 10.3390/antiox14030270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 04/15/2025] Open
Abstract
This study aimed to investigate the mechanisms by which large fibrous connective (LFC) tissue enhances fat graft survival in fat transplantation. A block fat graft model demonstrated that intact fat containing LFC showed significantly higher survival rates compared with liposuctioned fat. In the center of intact grafts, viable fat cells surrounded the LFC, forming a mesh-like living tissue structure. Proteomics of the extracellular matrix (ECM) adjacent to LFC (ALFC) and distant to LFC (DLFC) revealed significant differences in mitochondrial aspects. Staining of LFC tissue showed that it contains a large number of blood vessels and mitochondria, and exhibits stronger antioxidant capacity (p < 0.05) compared with adipose tissue. By mixing LFC with liposuctioned fat and transplanting into nude mice, histological sections showed that LFC promotes SOD1 expression, enhances respiratory chain RNA expression, and reduces ROS and inflammation. Pure mitochondrial-assisted fat transplantation only reduced short-term graft inflammation without improving long-term survival rates. In conclusion, LFC enhances long-term survival rates by reducing oxidative stress in fat grafts and forming a center for fat cell survival, thereby overcoming distance limitations. This represents a novel mechanism distinct from classical fat survival models and provides a reference for clinical practice.
Collapse
|
19
|
Chen M, Li L, Qin Y, Teng H, Lu C, Mai R, Zhu Z, Mo J, Qi Z. Mogroside V ameliorates astrocyte inflammation induced by cerebral ischemia through suppressing TLR4/TRADD pathway. Int Immunopharmacol 2025; 148:114085. [PMID: 39847949 DOI: 10.1016/j.intimp.2025.114085] [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: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
Inflammation and oxidative stress are pivotal factors in the onset and progression of secondary injury following cerebral ischemia-reperfusion (I/R). Mogroside V (MV), a primary active compound of Siraitia grosvenorii, exhibits significant anti-inflammatory and antioxidant properties. However, its specific effects in cerebral ischemia remain unclear. In this study, we evaluated the neuroprotective effects of MV in a model of focal cerebral ischemia. Male C57BL/6J mice were subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) as an in vivo model of cerebral ischemia-reperfusion injury (CIRI), while U87 cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to simulate CIRI in vitro. MV administration was found to reduce mortality, infarct volume, cerebral edema, and alleviate neurological deficits in these I/R mice. Furthermore, MV mitigated cerebral I/R injury by decreasing oxidative stress markers, such as reactive oxygen species (ROS) and malondialdehyde (MDA), while enhancing superoxide dismutase (SOD) levels. Gene Set Enrichment Analysis (GSEA) of the KEGG pathway revealed that most differentially expressed genes (DEGs) were involved in the Toll-like receptor/NF-κB/TNF/apoptosis signaling pathway. These findings were confirmed by real-time PCR, western blotting, immunohistochemistry, and immunofluorescence co-localization which demonstrated that MV reduced astrocyte inflammatory responses by inhibiting cytokine secretion associated with the TLR4/TRADD pathway. Additionally, MV protected neurons from apoptosis, as supported by TUNEL, Nissl, and HE staining. In conclusion, MV attenuates astrocyte inflammation and exerts neuroprotective effects following cerebral I/R injury, likely through suppression of the TLR4/TRADD signaling pathway.
Collapse
Affiliation(s)
- Meirong Chen
- Medical College of Guangxi University, Guangxi University, Nanning 530004, China; The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China
| | - Liangxian Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China
| | - Yang Qin
- Department of Graduate and Postgraduate Education Management, The Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Huanyao Teng
- School of Clinical Medicine, Guilin Medical University, Guilin 541199, China
| | - Chungui Lu
- School of Clinical Medicine, Guilin Medical University, Guilin 541199, China
| | - Ruyu Mai
- School of Clinical Medicine, Guilin Medical University, Guilin 541199, China
| | - Zhifei Zhu
- School of Clinical Medicine, Guilin Medical University, Guilin 541199, China
| | - Jingxin Mo
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Lab of Neurology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| | - Zhongquan Qi
- Medical College of Guangxi University, Guangxi University, Nanning 530004, China; Fujian Maternity and Child Health Hospital, Fuzhou, 350001, China; Stem Cell Therapy Research Center, Fuzhou 350001, China..
| |
Collapse
|
20
|
Zhao Z, Song H, Qi M, Liu Y, Zhang Y, Li S, Zhang H, Sun Y, Sun Y, Gao Z. Brain targeted polymeric micelles as drug carriers for ischaemic stroke treatment. J Drug Target 2025; 33:232-248. [PMID: 39403962 DOI: 10.1080/1061186x.2024.2417190] [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: 08/24/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke is a central nervous system disease with high morbidity, recurrence and mortality rates. Thrombolytic and neuroprotective therapies are the main therapeutic strategies for ischaemic stroke, however, the poor delivery efficiency of thrombolytic and neuroprotective drugs to the brain limits their clinical application. So far, the development of nanomedicine has brought opportunities for the above challenges, which can not only realise the effective accumulation of drugs in the target site, but also improve the pharmacokinetic behaviour of the drugs. Among the most rapidly developing nanoparticles, micelles gradually emerging as an effective strategy for ischaemic stroke treatment due to their own unique advantages. This review provided an overview of targeted and response-release micelles based on the physicochemical properties of the ischaemic stroke microenvironment, summarised the targeting strategies for delivering micellar formulations to the thrombus, blood-brain barrier, and brain parenchyma, and finally described the potentials and challenges of polymeric micelles in the treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Zirui Zhao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huijia Song
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Mengge Qi
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yurong Liu
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanchao Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Shuo Li
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huimin Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yongjun Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanping Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Zibin Gao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| |
Collapse
|
21
|
Li Y, Liu J, Pei D, Di D. Structural Characterization of, and Protective Effects Against, CoCl 2-Induced Hypoxia Injury to a Novel Neutral Polysaccharide from Lycium barbarum L. Foods 2025; 14:339. [PMID: 39941931 PMCID: PMC11818000 DOI: 10.3390/foods14030339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/17/2024] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Oxidative stress is closely related to the occurrence and development of ischaemic stroke. Natural plant polysaccharides have potential value in inhibiting oxidative stress and preventing ischaemic stroke. Here, a novel neutral polysaccharide named LICP009-3F-1a with a Mw of 10,780 Da was separated and purified from Lycium barbarum L. fruits. Linkage and NMR data revealed that LICP009-3F-1a has the following backbone: →4)-β-D-Glcp-(1→6)-β-D-Galp-(1→, with a branched chain of β-D-Galp-(1→3)-β-D-Galp-(1→, α-L-Araf-(1→ and →6)-α-D-Glcp-(1→ connected to the main chain through O-3 of →3,6)-β-D-Galp-(1→. X-ray and SEM analyses showed that LICP009-3F-1a has a semicrystalline structure with a laminar morphology. Thermal property analysis showed that LICP009-3F-1a is thermally stable. In vivo experiments suggested that LICP009-3F-1a could inhibit hypoxia-induced oxidative stress damage by eliminating ROS, reversing and restoring the activities of the antioxidant enzymes SOD, CAT, and GPx, and reducing the expression levels of the HIF-1α and VEGF genes. Blocking the apoptosis genes Bax and Caspase 3 and upregulating the expression of the antiapoptotic gene Bcl-2 protected PC12 cells from hypoxia-induced apoptosis. These results suggest that LICP009-3F-1a may have multiple potential uses in the treatment of IS.
Collapse
Affiliation(s)
| | | | | | - Duolong Di
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, No. 18, Tianshui Middle Road, Lanzhou 730000, China; (Y.L.); (J.L.); (D.P.)
| |
Collapse
|
22
|
Liu J, Zhao W, Kang J, Li X, Han L, Hu Z, Zhou J, Meng X, Gao X, Zhang Y, Gu Y, Liu X, Chen X. Halcinonide activates smoothened to ameliorate ischemic stroke injury. Life Sci 2025; 361:123324. [PMID: 39710062 DOI: 10.1016/j.lfs.2024.123324] [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: 08/23/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
OBJECTIVES The Shh pathway may shed new light on developing new cell death inhibitors for the therapy of ischemic stroke. We aimed to examine whether the Shh co-reporter SMO or its agonist halcinonide can upregulate Bcl-2 to suppress neuronal cell death, ultimately improving behavioral deficits and reducing cerebral infarction in an ischemic stroke model. METHODS Halcinonide or genetic manipulation of SMO was conducted in PC12 cells to examine their impacts on oxidative or OGD/R stress, and the chemical, along with AAV-SMO or AAV-EGFP were tested in MCAO rats to investigate their potential protective effects against neuronal damages due to cerebral I/R injury. The amounts or activities of L-LA, LDH, ROS, MDA, SOD, MPO, GSSG, and GSH were detected using the corresponding biochemical kits. The levels of TNF-α and IL-6 were analyzed by ELISA. RESULTS The results show that halcinonide alleviated neurological score and cerebral infarction, and the abnormal changes in L-LA, LDH, MDA, SOD, MPO, GSH, GSSG, TNF-α, and IL-6 were also reversed in MCAO rats. Through expression or knockout of SMO, we discovered that SMO worked similarly to halcinonide, protecting neuronal cells from oxidative or OGD/R stress, and AAV-SMO prevented cerebral damages of MCAO rats caused by ischemia and reperfusion. Halcinonide inhibited Bcl-2/Bax-mediated apoptosis, at least partially by promoting the Shh signaling pathway through enhancing SMO expression in vivo and in vitro. CONCLUSION This study identified a new target and a candidate chemical for therapy of ischemic stroke, hopefully reducing its morbidity and mortality.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; School of Basic Medical Sciences, University of South China, Hengyang, Hunan 421001, PR China.
| | - Wenyang Zhao
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Jia Kang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Xiangxiang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Liang Han
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Zhuozhou Hu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Jing Zhou
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Xinrui Meng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Xiaoshan Gao
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Yixuan Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Youquan Gu
- Department of Neurology, First Hospital of Lanzhou University, Lanzhou 730000, PR China.
| | - Xiaohua Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China.
| | - Xinping Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
23
|
Liu J, Han Y, Wu Z, Chen M, Wu W, Zhao Z, Yuan J, Zheng Z, Lin Q, Liu N, Chen H. Perillaldehyde pretreatment alleviates cerebral ischemia-reperfusion injury by improving mitochondrial structure and function via the Nrf2/Keap1/Trx2 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156328. [PMID: 39765034 DOI: 10.1016/j.phymed.2024.156328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 11/18/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Perilladehyde, an extract of perillae in the Labiatae family, can produce significant anti-inflammatory and antioxidant effects. Although literature evidences the favorable effect of perillaldehyde on ischemic stroke, the exact mechanism remains blurred. PURPOSE This study attempted to explore the impact of perillaldehyde on cerebral ischemia-reperfusion injury and the related action mechanism. METHODS The rat tMCAO and neuronal OGD/R models were established to simulate cerebral ischemia-reperfusion injury. Lentiviruses were used to interfere with the expression of Nrf2 and Trx2 in neurons. The effects and action mechanisms of perillaldehyde were explored by various experimental methods, including chromatin immunoprecipitation assay, Western Blot, flow cytometry, dual-luciferase reporter gene assay, transmission electron microscopy, MRI, RNA-seq, and immunofluorescence staining. RESULTS Perillaldehyde pretreatment effectively mitigated the tMCAO-induced brain injury in rats by reducing cerebral infarction, improving neuromotor function, and attenuating cell apoptosis in the ischemic penumbra. In vitro, perillaldehyde pretreatment alleviated cell death and excessive oxidative stress, preserved the mitochondrial membrane integrity, enhanced mitochondrial energy metabolism, and facilitated the restoration of mitochondrial ultrastructure after OGD/R. The mechanism probe revealed that perillaldehyde activated the Nrf2/Keap1/Trx2 signaling axis, thus promoting the transcription of Trx2 and improving mitochondrial structure and function. The aforementioned impacts of perillaldehyde were somewhat counteracted by disrupting the expression of Nrf2 and Trx2, suggesting that the neuroprotection of perillaldehyde partially involves the activation of the Nrf2/Keap1/Trx2 axis. CONCLUSIONS This study firstly demonstrates the existence of the Nrf2/Keap1/Trx2 signaling axis in cerebral ischemia-reperfusion injury and evidences that perillaldehyde pretreatment can promote the restoration of neuronal mitochondrial structure and function by activating the Nrf2/Keap1/Trx2 axis after cerebral ischemia-reperfusion injury. These findings signify that perillaldehyde holds great promises for clinical management of ischemic stroke.
Collapse
Affiliation(s)
- Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Ying Han
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhiyun Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Wenwen Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zijun Zhao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Jinjin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zhijian Zheng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Qiang Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
24
|
Shen L, Tian Q, Ran Q, Gan Q, Hu Y, Du D, Qin Z, Duan X, Zhu X, Huang W. Z-Ligustilide: A Potential Therapeutic Agent for Atherosclerosis Complicating Cerebrovascular Disease. Biomolecules 2024; 14:1623. [PMID: 39766330 PMCID: PMC11726876 DOI: 10.3390/biom14121623] [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: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis (AS) is one of the major catalysts of ischemic cerebrovascular disease, and the death and disease burden from AS and its cerebrovascular complications are increasing. Z-ligustilide (Z-LIG) is a key active ingredient in Angelica sinensis (Oliv.) Diels and Ligusticum chuanxiong Hort. In this paper, we first introduced LIG's physicochemical properties and pharmacokinetics. Then, we reviewed Z-LIG's intervention and therapeutic mechanisms on AS and its cerebrovascular complications. The mechanisms of Z-LIG intervention in AS include improving lipid metabolism, antioxidant and anti-inflammatory effects, protecting vascular endothelium, and inhibiting vascular endothelial fibrosis, pathological thickening, and plaque calcification. In ischemic cerebrovascular diseases complicated by AS, Z-LIG exerts practical neuroprotective effects in ischemic stroke (IS), transient ischemic attack (TIA), and vascular dementia (VaD) through anti-neuroinflammatory, anti-oxidation, anti-neuronal apoptosis, protection of the blood-brain barrier, promotion of mitochondrial division and angiogenesis, improvement of cholinergic activity, inhibition of astrocyte proliferation, and endoplasmic reticulum stress. This paper aims to provide a basis for subsequent studies of Z-LIG in the prevention and treatment of AS and its cerebrovascular complications and, thus, to promote the development of interventional drugs for AS.
Collapse
Affiliation(s)
- Longyu Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Qiqi Ran
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianrong Gan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Donglian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Zehua Qin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyi Duan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyun Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| |
Collapse
|
25
|
Song M, Zeng F, Huang L, Xiao H, Yi F, Sun X, Zheng L, Du H, Gu W. Energy restriction inhibits β-catenin ubiquitination to improve ischemic stroke injury via USP18/SKP2 axis. Metab Brain Dis 2024; 40:68. [PMID: 39692824 DOI: 10.1007/s11011-024-01494-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
Ischemic stroke (IS) remains a global health issue because of its great disability and mortality. Energy restriction (ER) has been justified to perform an inhibitory role in cerebral injury caused by IS. This research was purposed to inquire the potential molecular mechanism of ER in IS. To verify the function of ER in the animal and cell models of IS, rats were subjected to intermittent fasting (IF) and middle cerebral artery occlusion/reperfusion (MCAO/R) surgery and HAPI cells were treated with oxygen-glucose deprivation and reoxygenation (OGD/R) and 2-deoxyglucose (2-DG). It was disclosed that IF mitigated brain damage and inflammation in MCAO/R rats. Likewise, ER inhibited OGD/R-evoked microglial activation and inflammatory response. Of note, ubiquitin specific protease 18 (USP18) was uncovered to be the most significantly upregulated in MCAO/R rats receiving IF compared to free-feeding MCAO/R rats. Real-time quantitative polymerase chain reaction (RT-qPCR) and western blot verified that ER led to the promotion of USP18 expression. Moreover, downregulation of USP18 neutralized the meliorative effects of ER on OGD/R-treated HAPI cells. Functionally, USP18 restrained β-catenin ubiquitination to enhance its expression. In addition, our results manifested that S-phase kinase associated protein 2 (SKP2) contributed to degradation of β-catenin and USP18 abolished the role of SKP2 in β-catenin ubiquitination. Knockout of USP18 eliminated the protective effects of IF on MCAO/R rats, while SKP2 exacerbated brain damage and inflammation by decreasing β-catenin expression after IF. In summary, we validated that ER-induced USP18 exerts a suppressive function in IS damage through SKP2-mediated β-catenin ubiquitination.
Collapse
Affiliation(s)
- Mingyu Song
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
| | - Feiyue Zeng
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Hui Xiao
- Department of Neurology, Changsha Central Hospital, Changsha, Hunan, China
| | - Fang Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Geriatric neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinyu Sun
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
| | - Lan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
| | - Hao Du
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China.
| |
Collapse
|
26
|
Shimizu Y, Yamaji T, Inoue M, Yasuda N, Yamagishi K, Sawada N, Tsugane S, Iwasaki M. Plasma uric acid levels and risk of dementia in a population-based cohort study. J Neurol Sci 2024; 467:123323. [PMID: 39612638 DOI: 10.1016/j.jns.2024.123323] [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: 07/21/2024] [Revised: 10/23/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Given the opposing properties of uric acid (UA), which are intracellular prooxidant action and extracellular antioxidant action, the association of circulating UA levels with dementia remains controversial. We aimed to ascertain whether both lower and higher plasma UA levels are associated with the risk of incident dementia among middle-aged and older population. METHODS 1685 participants (530 men and 1155 women) aged 40-69 years at baseline (1990-1993) were randomly selected for plasma UA measurement from base cohort participants who responded to the baseline questionnaire and provided blood samples. They were followed for dementia (disabling dementia requiring care; hereinafter dementia) from 2006 to 2016 using certification records for national long-term care insurance in Japan. A Cox proportional hazards model adjusted for various lifestyle factors and past medical history (cardiometabolic disease) was applied for overall participants and sex. RESULTS Dementia was diagnosed in 240 participants (14.2 % overall: 16.0 % in men and 13.4 % in women). No statistically significant association with plasma UA was found in overall participants. Compared to participants with UA of 5.1-6.0 mg/dL, men with ≥6.1 mg/dL showed fully adjusted hazard ratios of 1.46 (95 % confidence interval: 0.78-2.75) for 6.1-7.0 mg/dL and 1.89 (0.97-3.66) for ≥7.1 mg/dL, while women with ≥6.1 mg/dL showed 1.13 (0.54-2.38). CONCLUSIONS No statistically significant association between plasma UA level and risk of dementia was found in overall participants or by sex.
Collapse
Affiliation(s)
- Yoko Shimizu
- Division of Epidemiology, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Taiki Yamaji
- Division of Epidemiology, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Manami Inoue
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Division of Prevention, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Nobufumi Yasuda
- Department of Public Health, Kochi University Medical School, 185-1 Kohasu, Oko-town, Nankoku-city, Kochi-prefecture, 783-8505, Japan.
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Institute of Medicine, and Health Services Research Center, University of Tsukuba, Tennodai 1-1-1, Tsukuba 305-8575, Japan; Department of Public Health, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Norie Sawada
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Shoichiro Tsugane
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; International University of Health and Welfare Graduate School of Public Health, 4-1-26 Akasaka, Minato City, Tokyo 107-8402, Japan.
| | - Motoki Iwasaki
- Division of Epidemiology, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Division of Cohort Research, National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
27
|
Kobzeva KA, Gurtovoy DE, Polonikov AV, Pokrovsky VM, Patrakhanov EA, Bushueva OY. Polymorphism in Genes Encoding HSP40 Family Proteins is Associated with Ischemic Stroke Risk and Brain Infarct Size: A Pilot Study. J Integr Neurosci 2024; 23:211. [PMID: 39735968 DOI: 10.31083/j.jin2312211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Heat shock proteins (HSPs) play a critical role in the molecular mechanisms of ischemic stroke (IS). A possible role for HSP40 family proteins in atherosclerosis progression has already been revealed; however, to date, molecular genetic studies on the involvement of genes encoding proteins of the HSP40 family in IS have not yet been carried out. AIM We sought to determine whether nine single nucleotide polymorphisms (SNPs) in genes encoding HSP40 family proteins (DNAJB1, DNAJB2, DNAJA1, DNAJA2, DNAJA3 and DNAJC7) are associated with the risk and clinical features of IS. METHODS Using TaqMan-based polymerase chain reaction (PCR) and the MassArray-4 system, DNA samples of 2551 Russians - 1306 IS patients and 1245 healthy individuals - were genotyped. RESULTS SNP rs2034598 DNAJA2 decreased the risk of IS exclusively in male patients (odds ratio = 0.81, 95% confidence interval 0.78-0.98, p = 0.028); rs7189628 DNAJA2 increased the brain infarct size (p = 0.04); and rs6500605 DNAJA3 lowered the age of onset of IS (p = 0.03). SNPs rs10448231 DNAJA1, rs7189628 DNAJA2, rs4926222 DNAJB1 and rs2034598 DNAJA2 were involved in the strongest epistatic interactions linked to IS; SNP rs10448231 DNAJA1 is characterised by the most essential mono-effect (2.96% of IS entropy); all of the top SNP-SNP interaction models included the pairwise combination rs7189628 DNAJA2×rs4926222 DNAJB1, which was found to be a key factor determining susceptibility to IS. In interactions with the studied SNPs, smoking was found to have multidirectional effects (synergism, antagonism or additive effect) and the strongest mono-effect (3.47% of IS entropy), exceeding the mono-effects of rs6500605 DNAJA3, rs10448231 DNAJA1, rs2034598 DNAJA2, rs7189628 DNAJA2 and rs4926222 DNAJB1, involved in the best G×E models and determining 0.03%-0.73% of IS entropy. CONCLUSIONS We are the first to discover polymorphisms in genes encoding HSP40 family proteins as a major risk factor for IS and its clinical manifestations. The comprehensive bioinformatics analysis revealed molecular mechanisms, underscoring their significance in the pathogenesis of IS, primarily reflecting the regulation of heat stress, proteostasis and cellular signalling.
Collapse
Affiliation(s)
- Ksenia A Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Denis E Gurtovoy
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Alexey V Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| | - Vladimir M Pokrovsky
- Laboratory of Genetic Technologies and Gene Editing for Biomedicine and Veterinary Medicine, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny A Patrakhanov
- Laboratory of Genetic Technologies and Gene Editing for Biomedicine and Veterinary Medicine, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olga Y Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| |
Collapse
|
28
|
Luo Y, Liu R, Yuan G, Pan Y. Polyphenols for stroke therapy: the role of oxidative stress regulation. Food Funct 2024; 15:11383-11399. [PMID: 39497601 DOI: 10.1039/d4fo01900h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Stroke is associated with a high incidence and disability rate, which seriously endangers human health. Oxidative stress (OS) plays a crucial role in the underlying pathologic progression of cerebral damage in stroke. Emerging experimental studies suggest that polyphenols have antioxidant potential and express protective effects after different types of strokes, but no breakthrough has been achieved in clinical studies. Nanomaterials, due to small characteristic sizes, can be used to deliver drugs, and have shown excellent performance in the treatment of various diseases. The drug delivery capability of nanomaterials has significant implications for the clinical translation and application of polyphenols. This comprehensive review introduces the mechanism of oxidative stress in stroke, and also summarizes the antioxidant effects of polyphenols on reactive oxygen species generation and oxidative stress after stroke. Also, the application characteristics and research progress of nanomaterials in the treatment of stroke with antioxidants are presented.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
29
|
Shao L, Wang C, Xu G, Tu Z, Yu X, Weng C, Liu J, Jian Z. Utilizing reactive oxygen species-scavenging nanoparticles for targeting oxidative stress in the treatment of ischemic stroke: A review. Open Med (Wars) 2024; 19:20241041. [PMID: 39588390 PMCID: PMC11587925 DOI: 10.1515/med-2024-1041] [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: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 11/27/2024] Open
Abstract
Ischemic stroke, which accounts for the majority of stroke cases, triggers a complex series of pathophysiological events, prominently characterized by acute oxidative stress due to excessive production of reactive oxygen species (ROS). Oxidative stress plays a crucial role in driving cell death and inflammation in ischemic stroke, making it a significant target for therapeutic intervention. Nanomedicine presents an innovative approach to directly mitigate oxidative damage. This review consolidates existing knowledge on the role of oxidative stress in ischemic stroke and assesses the potential of various ROS-scavenging nanoparticles (NPs) as therapeutic agents. We explore the properties and mechanisms of metal, metal-oxide, and carbon-based NPs, emphasizing their catalytic activity and biocompatibility in scavenging free radicals and facilitating the delivery of therapeutic agents across the blood-brain barrier. Additionally, we address the challenges such as cytotoxicity, immunogenicity, and biodistribution that need to be overcome to translate these nanotechnologies from bench to bedside. The future of NP-based therapies for ischemic stroke holds promise, with the potential to enhance outcomes through targeted modulation of oxidative stress.
Collapse
Affiliation(s)
- Lingmin Shao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Can Wang
- Department of Neurosurgery, Ezhou Central Hospital, Ezhou, 436000, Hubei, China
| | - Gang Xu
- Department of Neurosurgery, Xiantao First People’s Hospital, Xiantao, 433000, Hubei, China
| | - Zewei Tu
- Department of Neurosurgery, Yale School of Medicine, New Haven, 06510, CT, United States of America
| | - Xinyuan Yu
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, NC, United States of America
| | - Chao Weng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jia Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| |
Collapse
|
30
|
Wu G, Li B, Wei X, Chen Y, Zhao Y, Peng Y, Su J, Hu Z, Zhuo L, Tian Y, Wang Z, Peng X. Design, synthesis and biological evaluation of N-salicyloyl tryptamine derivatives as multifunctional neuroprotectants for the treatment of ischemic stroke. Eur J Med Chem 2024; 278:116795. [PMID: 39216381 DOI: 10.1016/j.ejmech.2024.116795] [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: 03/28/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke (IS) is a disease of high death and disability worldwide with few medications in clinical treatment. Neuroinflammation and oxidative stress are considered as crucial factors in the progression of IS. In our previous studies, N-salicyloyl tryptamine derivative (NST) L7 exhibited promising anti-inflammatory properties and is considered a potential clinical therapy for IS but had limited antioxidant capacity. Here, we have designed, synthesized, and biologically evaluated 30 novel NSTs for their neuroprotective effects against cerebral ischemia-reperfusion (CI/R) injury. To identify a multifunctional neuroprotectant with enhanced antioxidant and anti-inflammatory capacity, as well as an effective therapeutic agent for CI/R damage. Among them, M11 exhibited synergistic highly anti-oxidant, anti-inflammatory, anti-ferroptosis, and anti-apoptosis effects and surpassed the parent compound L7. Further studies demonstrated that the synergistic and efficient neuroprotective role of M11 was mainly achieved by activating Nrf2 and stimulating its downstream target HO-1/GCLC/NQO1/GPX4. In addition, M11 possessed good blood-brain barrier permeability. Moreover, M11 effectively reduced cerebral infarct volume and improved neurological deficits in MCAO/R mice. Its hydrochloride form, M11·HCl, exhibited better pharmacokinetic properties, high safety, and a significant reduction in infarct volume, which is comparable to Edaravone. In conclusion, our findings suggested that M11 capable of activating Nrf2, could represent a promising candidate agent for IS.
Collapse
Affiliation(s)
- Genping Wu
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Li
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiuzhen Wei
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yaxin Chen
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuting Zhao
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan Peng
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jianhui Su
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zecheng Hu
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linsheng Zhuo
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Tian
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Xue Peng
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
31
|
Yang Q, Li R, Hong Y, Liu H, Jian C, Zhao S. Curcumin-Loaded Gelatin Nanoparticles Cross the Blood-Brain Barrier to Treat Ischemic Stroke by Attenuating Oxidative Stress and Neuroinflammation. Int J Nanomedicine 2024; 19:11633-11649. [PMID: 39553455 PMCID: PMC11568047 DOI: 10.2147/ijn.s487628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024] Open
Abstract
Background Ischemic stroke is a medical emergency for which effective treatment remains inadequate. Curcumin (Cur) is a natural polyphenolic compound that is regarded as a potent neuroprotective agent. Compared to synthetic pharmaceuticals, Cur possesses minimal side effects and exhibits multiple mechanisms of action, offering significant advantages in the treatment of ischemic stroke. However, drawbacks such as poor water solubility and transmembrane permeability limit the efficacy of Cur. In recent years, nano-delivery systems have attracted great interest in the field of stroke therapy as an effective method to improve drug solubility and cross the blood-brain barrier (BBB). Methods In this study, a novel nanomedicine (Cur@GAR NPs) for ischemic stroke treatment was developed based on Cur-loaded gelatin nanoparticles (Cur@Gel NPs) that were then functionalized and modified with rabies virus glycoprotein (RVG29) to target brain tissue. The stability, antimicrobial properties, antioxidant properties, neuroprotective effects, neuronal cell uptake, and biocompatibility of Cur@GAR NPs were investigated in vitro. The in vivo therapeutic effect of Cur@GAR NPs on ischemic stroke was investigated in a middle cerebral artery occlusion (MCAO) rat model using the Morris water maze test and the open field test, and the potential mechanism of action was further investigated by histological analysis. Results The resulting Cur@GAR NPs improved the solubility of Cur and exhibited good dispersion. In vitro studies have shown that Cur@GAR NPs exhibit great antimicrobial properties, antioxidant properties and intracellular reactive oxygen species (ROS) protection. Notably, RVG29 significantly enhanced the uptake of Cur@GAR NPs by SH-SY5Y cells. Furthermore, in vivo studies verified the role of Cur@GAR NPs in reducing nerve damage and supporting neurological recovery. In the MCAO rat model, Cur@GAR NPs significantly attenuated neuroinflammation, reduced neuronal apoptosis and restored behavioral functions to a great extent. Conclusion Together these findings implied that Cur@GAR NPs could provide a novel and promising approach for effective ischemic stroke treatment.
Collapse
Affiliation(s)
- Qinglu Yang
- Department of Rehabilitation Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Ruitong Li
- Department of Psychology and Human Development, IOE, UCL’s Faculty of Education and Society, University College London, London, WC1H 0AL, UK
| | - Yigen Hong
- Department of Rehabilitation Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Hongsheng Liu
- Guangdong Huayi Biomedical Science and Technology Center, Guangzhou, Guangdong, People’s Republic of China
| | - Chuyao Jian
- Department of Rehabilitation Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Shaofeng Zhao
- Department of Rehabilitation Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
32
|
Wang Q, Li S, Wu W, Zhou W, Yan K, Liu Z, Yan L, Zheng B, Zhang F, Jiang X, Ye Y, Wang H. RNF13 protects neurons against ischemia-reperfusion injury via stabilizing p62-mediated Nrf2/HO-1 signaling pathway. Cell Commun Signal 2024; 22:535. [PMID: 39511649 PMCID: PMC11542339 DOI: 10.1186/s12964-024-01905-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Cerebral ischemia/reperfusion injury (CIRI), a common, universal clinical problem that costs a large proportion of the economic and disease burden. Identifying the key regulators of cerebral I/R injury could provide potential strategies for clinically improving the prognosis of stroke. Ring finger protein 13 (RNF13) has been proven to be involved in the inflammatory response. Here, we aimed to identify the role of RNF13 in cerebral I/R injury and further reveal its immanent mechanisms. METHODS The CRISPR/Cas9 based knockout mice, RNA sequencing, mass spectrometry, co-immunoprecipitation, GST-pull down, immunofluorescent staining, western blot, RT-PCR were used to investigate biodistribution, function and mechanism of RNF13 during cerebral I/R injury. RESULTS In the present study, we found that RNF13 was significantly up-regulated in patients, mice and primary neurons after I/R injury. Deficiency of RNF13 aggravated I/R-induced neurological impairment, inflammatory response and apoptosis while overexpression of RNF13 inhibited I/R injury. Mechanistically, this inhibitory effect of RNF13 during I/R injury was confirmed to be dependent on the blocking of TRIM21-mediated autophagy-dependent degradation of p62 and the stabilization of the p62-mediated Nrf2/HO-1 signaling pathway. CONCLUSION RNF13 is a crucial regulator of cerebral I/R injury that plays its role in inhibiting cell apoptosis and inflammatory response by preventing the autophagy-medicated degradation of the p62/Nrf2/HO-1 signaling pathway via blocking the interaction of TRIM21-p62 complex. Therefore, RNF13 represents a potential pharmacological target in acute ischemia stroke therapy.
Collapse
Affiliation(s)
- Qiangping Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Shuang Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wenke Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, China
| | - Kaixuan Yan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Zhen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Lanlan Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Baoping Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Fangcheng Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China.
| | - Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China.
| |
Collapse
|
33
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
34
|
Li T, Li H, Zhang S, Wang Y, He J, Kang J. Transcriptome Sequencing-Based Screening of Key Melatonin-Related Genes in Ischemic Stroke. Int J Mol Sci 2024; 25:11620. [PMID: 39519172 PMCID: PMC11547107 DOI: 10.3390/ijms252111620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemic stroke (IS) is a complex syndrome of neurological deficits due to stenosis or occlusion of the carotid and vertebral arteries for which there is still no effective treatment. Melatonin, a hormone secreted by the pineal gland, has multiple biological effects, such as antioxidant and anti-inflammatory properties, circadian rhythm regulation, and tissue regeneration, demonstrating potential applications in the treatment of IS. The aim of this study was to investigate key melatonin-regulated genes associated with IS using transcriptome sequencing and bioinformatics analyses and to explore their potential mechanisms of action in the disease process. We obtained gene expression data related to ischemic stroke (IS) from the Gene Expression Omnibus (GEO) database and identified candidate genes using machine learning algorithms. We then assessed the predictive power of these genes using PPI network analysis and diagnostic models. Finally, a series of enrichment analyses identified four key genes: ADM, PTGS2, MMP9, and VCAN. In addition, we determined the mRNA levels of these four key genes in an IS rat model using qPCR and found that all of these genes were significantly upregulated in the IS model compared to the control group, which is consistent with the results of previous analyses. Meanwhile, these genes have biological functions such as regulating vascular tone, participating in the inflammatory response, influencing tissue remodeling, and regulating cell adhesion and proliferation, playing key roles in the pathogenesis of IS. Therefore, we suggest that these four key genes may serve as prospective biomarkers for IS and help predict the risk of developing IS. In conclusion, this study elucidates for the first time the potential role of melatonin in the pathogenesis of IS and lays the foundation for in-depth studies on the functions of these key genes in the pathophysiology of IS and their potential applications in clinical diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jingsong Kang
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China; (T.L.); (H.L.); (S.Z.); (Y.W.); (J.H.)
| |
Collapse
|
35
|
Huang S, Zou S, Hee J, Gongye R, Xu S, Tang K. Pregnancy loss and risk of cardiometabolic multimorbidity in Chinese women: the China Kadoorie Biobank study. BMC Public Health 2024; 24:2694. [PMID: 39358758 PMCID: PMC11448432 DOI: 10.1186/s12889-024-20199-0] [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: 07/05/2023] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND While the association between pregnancy loss and individual cardiometabolic diseases (CMDs) is well-established, its impact on the risk of coexisting CMDs remains unclear. Therefore, the aim of this study is to investigate the association between pregnancy loss with the risk of cardiometabolic multimorbidity in Chinese women. METHOD We analyzed the cross-sectional data of 299,582 female participants aged 30-79 years old from the China Kadoorie biobank. Cardiometabolic multimorbidity was defined as the coexistence of two or more CMDs, including coronary heart disease, stroke, hypertension, and diabetes. Multivariable logistic regression was used to evaluate the odds ratios (ORs) between the number and type of pregnancy loss with the risk of cardiometabolic multimorbidity, characterized by the number and type of CMD. RESULTS After adjusting for confounding factors, pregnancy loss was found to be significantly associated with increased cardiometabolic multimorbidity risk (OR, 1.13 95% CI 1.08-1.19). Specifically, pregnancy loss due to spontaneous and induced abortion (OR 1.10, 95% CI 1.03-1.18 and OR 1.13, 95% CI 1.08-1.19, respectively). In contrast, no significant association was found between stillbirth and cardiometabolic multimorbidity (OR 1.03, 95% CI 0.95-1.11). The risk of cardiometabolic multimorbidity increases as the number of pregnancy loss increases (one pregnancy loss: OR 1.10, 95% CI 1.05-1.16, two or more pregnancy loss: OR 1.16, 95% CI 1.10-1.22). Similarly, the diagnosis of multiple CMDs increases with increasing number of pregnancy loss. Pregnancy loss was related to higher risk of cardiometabolic multimorbidity across most CMD combinations of CMDs. CONCLUSION Pregnancy loss, in particular, spontaneous and induced abortion was significantly associated with greater risk of cardiometabolic multimorbidity. The associations were stronger among those with recurrent pregnancy loss.
Collapse
Affiliation(s)
- Sha Huang
- School of Environmental Science and Engineering, Hainan University, Haikou, Hainan, 570228, China.
| | - Siyu Zou
- Vanke School of Public Health, Tsinghua University, Zhongguancun North Street, Haidian District, Beijing, 100084, China
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Maryland, USA
| | - Jiayi Hee
- Vanke School of Public Health, Tsinghua University, Zhongguancun North Street, Haidian District, Beijing, 100084, China
| | - Ruofan Gongye
- Vanke School of Public Health, Tsinghua University, Zhongguancun North Street, Haidian District, Beijing, 100084, China
| | - Shunqing Xu
- School of Environmental Science and Engineering, Hainan University, Haikou, Hainan, 570228, China.
| | - Kun Tang
- Vanke School of Public Health, Tsinghua University, Zhongguancun North Street, Haidian District, Beijing, 100084, China.
| |
Collapse
|
36
|
Zheng T, Jiang T, Ma H, Zhu Y, Wang M. Targeting PI3K/Akt in Cerebral Ischemia Reperfusion Injury Alleviation: From Signaling Networks to Targeted Therapy. Mol Neurobiol 2024; 61:7930-7949. [PMID: 38441860 DOI: 10.1007/s12035-024-04039-1] [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: 11/01/2023] [Accepted: 02/09/2024] [Indexed: 09/21/2024]
Abstract
Ischemia/reperfusion (I/R) injury is a pathological event that results in reperfusion due to low blood flow to an organ. Cerebral ischemia is a common cerebrovascular disease with high mortality, and reperfusion is the current standard intervention. However, reperfusion may further induce cellular damage and dysfunction known as cerebral ischemia/reperfusion injury (CIRI). Currently, strategies for the clinical management of CIRI are limited, necessitating the exploration of novel and efficacious treatment modalities for the benefit of patients. PI3K/Akt signaling pathway is an important cellular process associated with the disease. Stimulation of the PI3K/Akt pathway enhances I/R injury in multiple organs such as heart, brain, lung, and liver. It stands as a pivotal signaling pathway crucial for diminishing cerebral infarction size and safeguarding the functionality of brain tissue after CIRI. During CIRI, activation of the PI3K/Akt pathway exhibits a protective effect on CIRI. Furthermore, activation of the PI3K/Akt pathway has the potential to augment the activity of antioxidant enzymes, resulting in a decrease in reactive oxygen species (ROS) and the associated oxidative stress. Meanwhile, PI3K/Akt plays a neuroprotective role by inhibiting inflammatory responses and apoptosis. For example, PI3K/Akt interacts with NF-κB, Nrf2, and MAPK signaling pathways to mitigate CIRI. This article is aimed to explore the pivotal role and underlying mechanism of PI3K/Akt in ameliorating CIRI and investigate the influence of ischemic preconditioning and post-processing, as well as the impact of pertinent drugs or activators targeting the PI3K/Akt pathway on CIRI. The primary objective is to furnish compelling evidence supporting the activation of PI3K/Akt in the context of CIRI, elucidating its mechanistic intricacies. By doing so, the paper aims to underscore the critical contribution of PI3K/Akt in mitigating CIRI, providing a theoretical foundation for considering the PI3K/Akt pathway as a viable target for CIRI treatment.
Collapse
Affiliation(s)
- Ting Zheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Taotao Jiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Hongxiang Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Yanping Zhu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Manxia Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
37
|
Liang Y, Jiang Y, Liu J, Li X, Cheng X, Bao L, Zhou H, Guo Z. Blood-Brain Barrier Disruption and Imaging Assessment in Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01300-6. [PMID: 39322815 DOI: 10.1007/s12975-024-01300-6] [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: 07/09/2024] [Revised: 08/20/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Disruption of the blood-brain barrier (BBB) is an important pathological hallmark of ischemic stroke. Blood-brain barrier disruption (BBBD) is a consequence of ischemia and may also exacerbate damage to brain parenchyma. Therefore, maintaining BBB integrity is critical for the central nervous system (CNS) homeostasis. This review offers a concise overview of BBB structure and function, along with the mechanisms underlying its impairment following a stroke. In addition, we review the recent imaging techniques employed to study blood-brain barrier permeability (BBBP) in the context of ischemic brain injury with the goal of providing imaging guidance for stroke diagnosis and treatment from the perspective of the BBBD. This knowledge is vital for developing strategies to safeguard the BBB during cerebral ischemia.
Collapse
Affiliation(s)
- Yuchen Liang
- Department of Radiology, the First Hospital of Jilin University, Changchun, China
| | - Yueluan Jiang
- MR Research and Collaboration Team, Diagnostic Imaging, Siemens Healthineers Ltd., Beijing, China
| | - Jiaxin Liu
- Department of Radiology, the First Hospital of Jilin University, Changchun, China
| | - Xuewei Li
- Department of Radiology, the First Hospital of Jilin University, Changchun, China
| | - Xinyue Cheng
- Department of Radiology, the First Hospital of Jilin University, Changchun, China
| | - Lei Bao
- Department of Radiology, the First Hospital of Jilin University, Changchun, China
| | - Hongwei Zhou
- Department of Radiology, the First Hospital of Jilin University, Changchun, China.
| | - Zhenni Guo
- Department of Neurology, Stroke Center, the First Hospital of Jilin University, Changchun, China.
- Department of Neurology, Neuroscience Research Center, the First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
38
|
Zhu M, Wang Y, Park J, Titus A, Guo F. Dispensable regulation of brain development and myelination by the immune-related protein Serpina3n. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579239. [PMID: 38370831 PMCID: PMC10871299 DOI: 10.1101/2024.02.06.579239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Serine protease inhibitor clade A member 3n (Serpina3n) or its human orthologue SERPINA3 is a secretory immune-related molecule produced primarily in the liver and brain under homeostatic conditions and upregulated in response to system inflammation. Yet it remains elusive regarding its cellular identity and physiological significance in the development of the postnatal brain. Here, we reported that oligodendroglial lineage cells are the major cell population expressing Serpina3n protein in the postnatal murine CNS. Using loss-of-function genetic tools, we found that Serpina3n conditional knockout (cKO) from Olig2-expressing cells does not significantly affect cognitive and motor functions in mice. Serpina3n depletion does not appear to interfere with oligodendrocyte differentiation and developmental myelination nor affects the population of other glial cells and neurons in vivo. Together, these data suggest that the immune-related molecule Serpina3n plays a minor role, if any, in regulating neural cell development in the postnatal brain under homeostatic conditions. We found that Serpina3n is significantly upregulated in response to oxidative stress, and it potentiates oxidative injury and cell senescence of oligodendrocytes. Our data raise the interest in pursuing its functional significance in the CNS under disease/injury conditions.
Collapse
Affiliation(s)
- Meina Zhu
- Department of Neurology, School of Medicine, UC Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA
| | - Yan Wang
- Department of Neurology, School of Medicine, UC Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA
| | - Joohyun Park
- Department of Neurology, School of Medicine, UC Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA
| | - Annlin Titus
- Department of Neurology, School of Medicine, UC Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA
| | - Fuzheng Guo
- Department of Neurology, School of Medicine, UC Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA
| |
Collapse
|
39
|
Zhao B, Li M, Li B, Li Y, Shen Q, Hou J, Wu Y, Gu L, Gao W. The action mechanism by which C1q/tumor necrosis factor-related protein-6 alleviates cerebral ischemia/reperfusion injury in diabetic mice. Neural Regen Res 2024; 19:2019-2026. [PMID: 38227531 DOI: 10.4103/1673-5374.390951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 07/29/2023] [Indexed: 01/17/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202409000-00034/figure1/v/2024-01-16T170235Z/r/image-tiff Studies have shown that C1q/tumor necrosis factor-related protein-6 (CTRP6) can alleviate renal ischemia/reperfusion injury in mice. However, its role in the brain remains poorly understood. To investigate the role of CTRP6 in cerebral ischemia/reperfusion injury associated with diabetes mellitus, a diabetes mellitus mouse model of cerebral ischemia/reperfusion injury was established by occlusion of the middle cerebral artery. To overexpress CTRP6 in the brain, an adeno-associated virus carrying CTRP6 was injected into the lateral ventricle. The result was that oxygen injury and inflammation in brain tissue were clearly attenuated, and the number of neurons was greatly reduced. In vitro experiments showed that CTRP6 knockout exacerbated oxidative damage, inflammatory reaction, and apoptosis in cerebral cortical neurons in high glucose hypoxia-simulated diabetic cerebral ischemia/reperfusion injury. CTRP6 overexpression enhanced the sirtuin-1 signaling pathway in diabetic brains after ischemia/reperfusion injury. To investigate the mechanism underlying these effects, we examined mice with depletion of brain tissue-specific sirtuin-1. CTRP6-like protection was achieved by activating the sirtuin-1 signaling pathway. Taken together, these results indicate that CTRP6 likely attenuates cerebral ischemia/reperfusion injury through activation of the sirtuin-1 signaling pathway.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Mei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Bingyu Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yanan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qianni Shen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
40
|
Yu J, Wu M, Shi M, Gong Y, Gao F, Gu H, Dang B. Up-regulation of BMAL1 by epigallocatechin-3-gallate improves neurological damage in SBI rats. Brain Res Bull 2024; 215:111033. [PMID: 39032586 DOI: 10.1016/j.brainresbull.2024.111033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Brain Muscle ARNT-Like Protein 1 (BMAL1) suppresses oxidative stress in brain injury during surgery. Epigallocatechin-3-gallate (EGCG), a monomer in green tea, has been identified as an antioxidant and a potential agonist for BMAL1. In this work, the mechanism by which BMAL1 is regulated was investigated, as well as the therapeutic effect of EGCG on surgically injured rats. The pathological environment after brain injury during surgery was simulated by excising the right frontal lobe of rats. Rats received an intraperitoneal injection of EGCG immediately after surgery. Neurological scores and cerebral edema were recorded after surgery. Fluoro-Jade C staining, TUNEL staining, western blot, and lipid peroxidation analyses were conducted 3 days later. Here we show that the endogenous BMAL1 level decreased after brain injury. Postoperative administration of EGCG up-regulated the content of BMAL1 around the cerebral cortex, reduced the oxidative stress level, reduced neuronal apoptosis and the number of degenerated neurons, alleviated cerebral edema, and improved neurological scores in rats. This suggests that BMAL1 is an effective target for treating surgical brain injury, as well as that EGCG may be a promising agent for alleviating postoperative brain injury.
Collapse
Affiliation(s)
- Jiejie Yu
- Department of Emergency, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Muyao Wu
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Mengying Shi
- Department of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Yating Gong
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Fan Gao
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Haiping Gu
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Baoqi Dang
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China.
| |
Collapse
|
41
|
Chu CS, Chen YT, Liang WZ. Investigation of the mechanisms behind ochratoxin A-induced cytotoxicity in human astrocytes and the protective effects of N-acetylcysteine. J Appl Toxicol 2024; 44:1454-1465. [PMID: 38812125 DOI: 10.1002/jat.4652] [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: 03/14/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Ochratoxin A (OTA) is a type of mycotoxin commonly found in raw and processed foods. It is essential to be aware of this toxin, as it can harm your health if consumed in high quantities. OTA can induce toxic effects in various cell models. However, a more comprehensive understanding of the harmful effects of OTA on human astrocytes is required. This study evaluated OTA's neurotoxic effects on the Gibco® Human Astrocyte (GHA) cell line, its underlying mechanisms, and the antioxidant N-acetylcysteine (NAC) ability to prevent them. OTA exposure within 5-30 μM has induced concentration-dependent cytotoxicity. In the OTA-treated cells, the levels of reactive oxygen species (ROS) were found to be significantly increased, while the glutathione (GSH) contents were found to decrease considerably. The western blotting of OTA-treated cells has revealed increased Bax, cleaved caspase-9/caspase-3 protein levels, and increased Bax/Bcl-2 ratio. In addition, exposure to OTA has resulted in the induction of antioxidant responses associated with the protein expressions of Nrf2, HO-1, and NQO1. On the other hand, the pretreatment with NAC has partially alleviated the significant toxic effects of OTA. In conclusion, our findings suggest that oxidative stress and apoptosis are involved in the OTA-induced cytotoxicity in GHA cells. NAC could act as a protective agent against OTA-induced oxidative damage.
Collapse
Affiliation(s)
- Che-Sheng Chu
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ying-Tso Chen
- Department of Neurosurgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Zhe Liang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, Taiwan
| |
Collapse
|
42
|
Capobianco DL, De Zio R, Profico DC, Gelati M, Simone L, D'Erchia AM, Di Palma F, Mormone E, Bernardi P, Sbarbati A, Gerbino A, Pesole G, Vescovi AL, Svelto M, Pisani F. Human neural stem cells derived from fetal human brain communicate with each other and rescue ischemic neuronal cells through tunneling nanotubes. Cell Death Dis 2024; 15:639. [PMID: 39217148 PMCID: PMC11365985 DOI: 10.1038/s41419-024-07005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Pre-clinical trials have demonstrated the neuroprotective effects of transplanted human neural stem cells (hNSCs) during the post-ischemic phase. However, the exact neuroprotective mechanism remains unclear. Tunneling nanotubes (TNTs) are long plasma membrane bridges that physically connect distant cells, enabling the intercellular transfer of mitochondria and contributing to post-ischemic repair processes. Whether hNSCs communicate through TNTs and their role in post-ischemic neuroprotection remains unknown. In this study, non-immortalized hNSC lines derived from fetal human brain tissues were examined to explore these possibilities and assess the post-ischemic neuroprotection potential of these hNSCs. Using Tau-STED super-resolution confocal microscopy, live cell time-lapse fluorescence microscopy, electron microscopy, and direct or non-contact homotypic co-cultures, we demonstrated that hNSCs generate nestin-positive TNTs in both 3D neurospheres and 2D cultures, through which they transfer functional mitochondria. Co-culturing hNSCs with differentiated SH-SY5Y (dSH-SY5Y) revealed heterotypic TNTs allowing mitochondrial transfer from hNSCs to dSH-SY5Y. To investigate the role of heterotypic TNTs in post-ischemic neuroprotection, dSH-SY5Y were subjected to oxygen-glucose deprivation (OGD) followed by reoxygenation (OGD/R) with or without hNSCs in direct or non-contact co-cultures. Compared to normoxia, OGD/R dSH-SY5Y became apoptotic with impaired electrical activity. When OGD/R dSH-SY5Y were co-cultured in direct contact with hNSCs, heterotypic TNTs enabled the transfer of functional mitochondria from hNSCs to OGD/R dSH-SY5Y, rescuing them from apoptosis and restoring the bioelectrical profile toward normoxic dSH-SY5Y. This complete neuroprotection did not occur in the non-contact co-culture. In summary, our data reveal the presence of a functional TNTs network containing nestin within hNSCs, demonstrate the involvement of TNTs in post-ischemic neuroprotection mediated by hNSCs, and highlight the strong efficacy of our hNSC lines in post-ischemic neuroprotection. Human neural stem cells (hNSCs) communicate with each other and rescue ischemic neurons through nestin-positive tunneling nanotubes (TNTs). A Functional mitochondria are exchanged via TNTs between hNSCs. B hNSCs transfer functional mitochondria to ischemic neurons through TNTs, rescuing neurons from ischemia/reperfusion ROS-dependent apoptosis.
Collapse
Affiliation(s)
- D L Capobianco
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - R De Zio
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - D C Profico
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni, Rotondo, Foggia, Italy
| | - M Gelati
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni, Rotondo, Foggia, Italy
| | - L Simone
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni, Rotondo, Foggia, Italy
| | - A M D'Erchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM) CNR, Bari, Italy
| | - F Di Palma
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - E Mormone
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni, Rotondo, Foggia, Italy
| | - P Bernardi
- Department of Neurosciences, Biomedicine and Movement Sciences. Unit of Human Anatomy, University of Verona, Verona, Italy
| | - A Sbarbati
- Department of Neurosciences, Biomedicine and Movement Sciences. Unit of Human Anatomy, University of Verona, Verona, Italy
| | - A Gerbino
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - G Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM) CNR, Bari, Italy
| | - A L Vescovi
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni, Rotondo, Foggia, Italy
- Faculty of Medicine, Link Campus University, Rome, Italy
| | - M Svelto
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM) CNR, Bari, Italy
- National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | - F Pisani
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy.
| |
Collapse
|
43
|
Jiao Y, Zhang X, Yang Z. SUMO-specific proteases: SENPs in oxidative stress-related signaling and diseases. Biofactors 2024; 50:910-921. [PMID: 38551331 DOI: 10.1002/biof.2055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/17/2024] [Indexed: 10/04/2024]
Abstract
Oxidative stress is employed to depict a series of responses detrimental to normal cellular functions resulting from an imbalance between intracellular oxidants, mainly reactive oxygen species and antioxidant defenses. Oxidative stress often contributes to the development of various diseases, including cancer, cardiovascular diseases, and neurodegenerative diseases. In this process, the relationship between small ubiquitin-like modifier (SUMO) and oxidative stress has garnered significant attention, with its posttranslational modification (PTM) frequently serving as a marker of oxidative stress status. Sentrin/SUMO-specific proteases (SENPs), affected by alternative splicing, PTMs such as phosphorylation and ubiquitination, and various protein interactions, are crucial molecules in the SUMO process. The human SENP family has six members (SENP1-3, SENP5-7), which are classified into two categories based on sequence similarity, substrate specificity, and subcellular location. They have two core functions in the human body: first, by cleaving the precursor SUMO and exposing the C-terminal glycine, they initiate the SUMO process; second, they can specifically recognize and dissociate SUMO proteins bound to substrates, a process known as deSUMOylation. However, the connection between deSUMOylation and oxidative stress remains a relatively unexplored area despite their strong association with oxidative diseases such as cancer and cardiovascular disease. This article aims to illustrate the significant contribution of SENPs to the oxidative stress pathway through deSUMOylation by reviewing their structure and classification, their roles in oxidative stress, and the changes in their expression and activity in several typical oxidative stress-related diseases.
Collapse
Affiliation(s)
- Yaqi Jiao
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Xiaojuan Zhang
- Department of Cell Biochemistry, University of Groningen, Groningen, The Netherlands
| | - Zhenshan Yang
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
44
|
Manoharan D, Wang LC, Chen YC, Li WP, Yeh CS. Catalytic Nanoparticles in Biomedical Applications: Exploiting Advanced Nanozymes for Therapeutics and Diagnostics. Adv Healthc Mater 2024; 13:e2400746. [PMID: 38683107 DOI: 10.1002/adhm.202400746] [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: 02/26/2024] [Revised: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Catalytic nanoparticles (CNPs) as heterogeneous catalyst reveals superior activity due to their physio-chemical features, such as high surface-to-volume ratio and unique optical, electric, and magnetic properties. The CNPs, based on their physio-chemical nature, can either increase the reactive oxygen species (ROS) level for tumor and antibacterial therapy or eliminate the ROS for cytoprotection, anti-inflammation, and anti-aging. In addition, the catalytic activity of nanozymes can specifically trigger a specific reaction accompanied by the optical feature change, presenting the feasibility of biosensor and bioimaging applications. Undoubtedly, CNPs play a pivotal role in pushing the evolution of technologies in medical and clinical fields, and advanced strategies and nanomaterials rely on the input of chemical experts to develop. Herein, a systematic and comprehensive review of the challenges and recent development of CNPs for biomedical applications is presented from the viewpoint of advanced nanomaterial with unique catalytic activity and additional functions. Furthermore, the biosafety issue of applying biodegradable and non-biodegradable nanozymes and future perspectives are critically discussed to guide a promising direction in developing span-new nanozymes and more intelligent strategies for overcoming the current clinical limitations.
Collapse
Affiliation(s)
- Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Liu-Chun Wang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Peng Li
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
45
|
Nabipour H, Rohani S. Metal-Organic Frameworks for Overcoming the Blood-Brain Barrier in the Treatment of Brain Diseases: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1379. [PMID: 39269041 PMCID: PMC11397546 DOI: 10.3390/nano14171379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024]
Abstract
The blood-brain barrier (BBB) plays a vital role in safeguarding the central nervous system by selectively controlling the movement of substances between the bloodstream and the brain, presenting a substantial obstacle for the administration of therapeutic agents to the brain. Recent breakthroughs in nanoparticle-based delivery systems, particularly metal-organic frameworks (MOFs), provide promising solutions for addressing the BBB. MOFs have become valuable tools in delivering medications to the brain with their ability to efficiently load drugs, release them over time, and modify their surface properties. This review focuses on the recent advancements in molecular-based approaches for treating brain disorders, such as glioblastoma multiforme, stroke, Parkinson's disease, and Alzheimer's disease. This paper highlights the significant impact of MOFs in overcoming the shortcomings of conventional brain drug delivery techniques and provides valuable insights for future research in the field of neurotherapeutics.
Collapse
Affiliation(s)
- Hafezeh Nabipour
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada
| | - Sohrab Rohani
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada
| |
Collapse
|
46
|
Shin JW, Jung KJ, Ryu M, Kim J, Kimm H, Jee SH. Causal association between serum bilirubin and ischemic stroke: multivariable Mendelian randomization. Epidemiol Health 2024; 46:e2024070. [PMID: 39210787 PMCID: PMC11826012 DOI: 10.4178/epih.e2024070] [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: 03/28/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES Previous research has predominantly focused on total bilirubin levels without clearly distinguishing between direct and indirect bilirubin. In this study, the differences between these forms were examined, and their potential causal relationships with ischemic stroke were investigated. METHODS Two-sample multivariable Mendelian randomization (MVMR) analysis was employed, extracting summary data on bilirubin from the Korean Cancer Prevention Study-II (n=159,844) and the Korean Genome and Epidemiology Study (n=72,299). Data on ischemic stroke were obtained from BioBank Japan (n=201,800). Colocalization analysis was performed, focusing on the UGT1A1, SLCO1B1, and SLCO1B3 genes, which are the primary loci associated with serum bilirubin levels. RESULTS Crude 2-sample Mendelian randomization analysis revealed a significant negative association between total bilirubin levels and ischemic stroke. However, in MVMR analyses, only indirect bilirubin demonstrated a significant negative association with ischemic stroke (odds ratio, 0.76; 95% confidence interval, 0.59 to 0.98). Colocalization analysis did not identify a shared causal variant between the 3 genetic loci related to indirect bilirubin and the risk of ischemic stroke. CONCLUSIONS Our study establishes a causal association between higher genetically determined levels of serum indirect bilirubin and reduced risk of ischemic stroke in an Asian population. Future research should include more in-depth analysis of shared genetic variants between indirect bilirubin and ischemic stroke.
Collapse
Affiliation(s)
- Jong Won Shin
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Ulsan, Korea
| | - Keum Ji Jung
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Mikyung Ryu
- Institute on Aging, Ajou University Medical Center, Suwon, Korea
- Basgenbio, Inc., Seoul, Korea
| | | | - Heejin Kimm
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| |
Collapse
|
47
|
Khan S, Bano N, Ahamad S, John U, Dar NJ, Bhat SA. Excitotoxicity, Oxytosis/Ferroptosis, and Neurodegeneration: Emerging Insights into Mitochondrial Mechanisms. Aging Dis 2024:AD.2024.0125-1. [PMID: 39122453 DOI: 10.14336/ad.2024.0125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in the development of age-related diseases, particularly neurodegenerative disorders. The etiology of mitochondrial dysfunction involves a multitude of factors that remain elusive. This review centers on elucidating the role(s) of excitotoxicity, oxytosis/ferroptosis and neurodegeneration within the context of mitochondrial bioenergetics, biogenesis, mitophagy and oxidative stress and explores their intricate interplay in the pathogenesis of neurodegenerative diseases. The effective coordination of mitochondrial turnover processes, notably mitophagy and biogenesis, is assumed to be critically important for cellular resilience and longevity. However, the age-associated decrease in mitophagy impedes the elimination of dysfunctional mitochondria, consequently impairing mitochondrial biogenesis. This deleterious cascade results in the accumulation of damaged mitochondria and deterioration of cellular functions. Both excitotoxicity and oxytosis/ferroptosis have been demonstrated to contribute significantly to the pathophysiology of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS) and Multiple Sclerosis (MS). Excitotoxicity, characterized by excessive glutamate signaling, initiates a cascade of events involving calcium dysregulation, energy depletion, and oxidative stress and is intricately linked to mitochondrial dysfunction. Furthermore, emerging concepts surrounding oxytosis/ferroptosis underscore the importance of iron-dependent lipid peroxidation and mitochondrial engagement in the pathogenesis of neurodegeneration. This review not only discusses the individual contributions of excitotoxicity and ferroptosis but also emphasizes their convergence with mitochondrial dysfunction, a key driver of neurodegenerative diseases. Understanding the intricate crosstalk between excitotoxicity, oxytosis/ferroptosis, and mitochondrial dysfunction holds potential to pave the way for mitochondrion-targeted therapeutic strategies. Such strategies, with a focus on bioenergetics, biogenesis, mitophagy, and oxidative stress, emerge as promising avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh-202002, India
| | - Urmilla John
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India; School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | | |
Collapse
|
48
|
He Y, Jin W, Wan H, Zhang L, Yu L. Research progress on immune-related therapeutic targets of brain injury caused by cerebral ischemia. Cytokine 2024; 180:156651. [PMID: 38761715 DOI: 10.1016/j.cyto.2024.156651] [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: 03/08/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Stroke is the second leading cause of death worldwide and a leading cause of disability. The innate immune response occurs immediately after cerebral ischemia, resulting in adaptive immunity. More and more experimental evidence has proved that the immune response caused by cerebral ischemia plays an important role in early brain injury and later the recovery of brain injury. Innate immune cells and adaptive cells promote the occurrence of cerebral ischemic injury but also protect brain cells. A large number of studies have shown that cytokines and immune-related substances also have dual functions of promoting injury, reducing injury, or promoting injury recovery in the later stage of cerebral ischemia. They can be an important target for treating cerebral ischemic recovery. Therefore, this study discussed the immune cells, cytokines, and immune-related substances with dual roles in cerebral ischemia and summarized the therapeutic targets of cerebral ischemia. To explore more effective methods to treat cerebral ischemia, promote the recovery of brain function, and improve the prognosis of patients.
Collapse
Affiliation(s)
- Yuejia He
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Lijiang Zhang
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Li Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
49
|
Kumari S, Dhapola R, Sharma P, Nagar P, Medhi B, HariKrishnaReddy D. The impact of cytokines in neuroinflammation-mediated stroke. Cytokine Growth Factor Rev 2024; 78:105-119. [PMID: 39004599 DOI: 10.1016/j.cytogfr.2024.06.002] [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: 04/13/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Cerebral stroke is ranked as the third most common contributor to global mortality and disability. The involvement of inflammatory mechanisms, both peripherally and within the CNS, holds significance in the pathophysiological cascades following the initiation of stroke. After the onset of acute stroke, predominantly ischemic, a subsequent phase of neuroinflammation ensues. It is a dual-effect process that not only exacerbates injury, leading to cell death, but paradoxically, it also serves a shielding role in facilitating recovery. Cytokines serve as pivotal mediators within the inflammatory cascade, actively contributing to the progression of ischemic damage. Stroke is followed by increased expression of pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, etc. leading to the recruitment and stimulation of glial cells and peripheral leukocytes at the site of injury, promoting neuroinflammation. Cytokines can directly induce neuronal injury and death through various mechanisms, including excitotoxicity, oxidative stress, HPA-axis activation, secretion of matrix metalloproteinase and apoptosis. They can also amplify the inflammatory response, leading to further neuronal damage. Therapeutic strategies aimed at modulating cytokine release, immune response and cytokine signalling or activity are being explored as potential interventions to mitigate neuroinflammation and its detrimental effects in stroke. In this review, we have given a concise summary of our current knowledge of the function of various cytokines, brain inflammation and various signalling and molecular pathways including JAK/STAT3, TGF-β/Smad, MAPK, HMGB1/TLR and NF-κB modulated cytokines regulation in stroke. Therapeutic agents such as MCC950, genistein, edaravone, minocycline, etc. targeting various cytokines-associated signalling pathways have shown efficacy in preclinical and clinical trials reducing the pathophysiology of the illness were also addressed in this study.
Collapse
Affiliation(s)
- Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Pushank Nagar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
50
|
Owjfard M, Rahimian Z, Karimi F, Borhani-Haghighi A, Mallahzadeh A. A comprehensive review on the neuroprotective potential of resveratrol in ischemic stroke. Heliyon 2024; 10:e34121. [PMID: 39082038 PMCID: PMC11284444 DOI: 10.1016/j.heliyon.2024.e34121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 06/07/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Stroke is the second leading cause of death and the third leading cause of disability worldwide. Globally, 68 % of all strokes are ischemic, with 32 % being hemorrhagic. Ischemic stroke (IS) poses significant challenges globally, necessitating the development of effective therapeutic strategies. IS is among the deadliest illnesses. Major functions are played by neuroimmunity, inflammation, and oxidative stress in the multiple intricate pathways of IS. Secondary brain damage is specifically caused by the early pro-inflammatory activity that follows cerebral ischemia, which is brought on by excessive activation of local microglia and the infiltration of circulating monocytes and macrophages. Resveratrol, a natural polyphenol found in grapes and berries, has shown promise as a neuroprotective agent in IS. This review offers a comprehensive overview of resveratrol's neuroprotective role in IS, focusing on its mechanisms of action and therapeutic potential. Resveratrol exerts neuroprotective effects by activating nuclear factor erythroid 2-related factor 2 (NRF2) and sirtuin 1 (SIRT1) pathways. SIRT1 activation by resveratrol triggers the deacetylation and activation of downstream targets like peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) and forkhead box protein O (FOXO), regulating mitochondrial biogenesis, antioxidant defense, and cellular stress response. Consequently, resveratrol promotes cellular survival and inhibits apoptosis in IS. Moreover, resveratrol activates the NRF2 pathway, a key mediator of the cellular antioxidant response. Activation of NRF2 through resveratrol enhances the expression of antioxidant enzymes, like heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1), which neutralize reactive oxygen species and mitigate oxidative stress in the ischemic brain. Combined, the activation of SIRT1 and NRF2 pathways contributes to resveratrol's neuroprotective effects by reducing oxidative stress, inflammation, and apoptosis in IS. Preclinical studies demonstrate that resveratrol improves functional outcomes, reduces infarct size, regulates cerebral blood flow and preserves neuronal integrity. Gaining a comprehensive understanding of these mechanisms holds promise for the development of targeted therapeutic interventions aimed at promoting neuronal survival and facilitating functional recovery in IS patients and to aid future studies in this matter.
Collapse
Affiliation(s)
- Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rahimian
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Arashk Mallahzadeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|