1
|
Zhang P, Ran Y, Han L, Li Y, Tian W, Sun X, Jiao M, Jing L, Luo X. Nanomaterial technologies for precision diagnosis and treatment of brain hemorrhage. Biomaterials 2025; 321:123269. [PMID: 40174300 DOI: 10.1016/j.biomaterials.2025.123269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Brain hemorrhage events present complex clinical challenges due to their rapid progression and the intricate interplay of oxidative stress, inflammation, and neuronal damage. Traditional diagnostic and therapeutic approaches often struggle to meet the demands for timely and effective intervention. This review explores the cutting-edge role of nanomaterials in transforming cerebral hemorrhage management, focusing on both diagnostic and therapeutic advancements. Nanomaterial-enabled imaging techniques, such as optical imaging, magnetic resonance imaging, and magnetic particle imaging, significantly enhance the accuracy of hemorrhage detection by providing real-time, high-resolution assessments of blood-brain barrier (BBB) integrity, cerebral perfusion, and hemorrhage progression, which is critical for guiding intervention strategies. On the therapeutic front, nanomaterial-based systems enable the precise delivery of drugs and bioactive molecules, fostering neural repair and functional recovery while minimizing systemic side effects. Furthermore, multifunctional nanomaterials not only address the primary injury but also offer precise control over secondary injuries, such as edema and oxidative stress. Their ability to enhance neuroprotection, prevent re-bleeding, and stimulate brain tissue regeneration provides a holistic approach and marks a significant advancement in brain hemorrhage therapy. As the field continues to advance, nanotechnology is set to fundamentally reshape the clinical management and long-term outcomes of brain hemorrhages, presenting a paradigm shift towards personalized and highly effective neurological care.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Yi'an Ran
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Lei Han
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Yao Li
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Wanru Tian
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Xiao Sun
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Mingxia Jiao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China.
| | - Lihong Jing
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.
| | - Xiliang Luo
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China.
| |
Collapse
|
2
|
Li H, Li X, Li M, Li W, Wei J, Huang Y, Yan H, Lin J, Zhang P. Edaravone dexborneol provides neuroprotective benefits by suppressing ferroptosis in experimental intracerebral hemorrhage. Sci Rep 2025; 15:16595. [PMID: 40360664 PMCID: PMC12075699 DOI: 10.1038/s41598-025-99187-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Edaravone dexborneol (EDB) is widely recognized for its anti-inflammatory and antioxidant properties and is clinically applied in the treatment of acute cerebral infarction. Ferroptosis is a critical process in the pathophysiology of brain injury following intracerebral hemorrhage (ICH). However, it remains unclear whether EDB can ameliorate ICH through the modulation of ferroptosis. This study aimed to evaluate the function and mechanism of EDB in treatment of ICH. With a male rat ICH model, animal behavior tests, histopathological staining, magnetic resonance imaging and evans blue staining were used to evaluate the neural protective function of EDB on ICH rats. The potential molecular mechanism was investigated using RNA sequencing. With the administration of Fer-1, a range of ferroptosis-related biomarkers, including Fe2+, 4-hydroxynonenal, malondialdehyde, etc., were analyzed to ascertain whether EDB confers neuroprotective effects through the modulation of P53/GPX4 pathways to inhibit ferroptosis. Finally, the findings were further corroborated using an in vitro ICH model with a P53 inhibitor. EDB has the potential to markedly enhance nerve and motor function, mitigate pathological damage, facilitate hematoma clearance, and repair BBB injury in ICH rats. KEGG analysis revealed that the differentially expressed genes were associated with signaling pathways, including P53 and ferroptosis. Both EDB and Fer-1 substantially reduced the concentrations of Fe2+, 4-hydroxynonenal, malondialdehyde, increased the amount of anti-oxidants, decreased the expression of P53, and concurrently upregulated the expression of GPX4. Besides, the P53 inhibitor PFT-α was observed to significantly reduce the levels of 4-HNE and lipid peroxides, while concurrently increasing the expression of GPX4. This investigation has shed light on the crucial neuroprotective role of EDB by regulating ferroptosis in ICH disease, which provided a theoretical basis for the clinical application of EDB in the treatment of ICH.
Collapse
Affiliation(s)
- Han Li
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiang Li
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
- Henan Department of Neurology, Henan Key Laboratory of Neural Regeneration and Repairment, Xinxiang, China
| | - Mingzhi Li
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Wenxin Li
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
| | - Jinghui Wei
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuming Huang
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Haiqing Yan
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China.
| |
Collapse
|
3
|
Franklin ME, Grant JL, Lee GM, Alvarez-Ciara A, Bennett C, Mattis S, Gallardo N, Corrales N, Cui XT, Capadona JR, Streit WJ, Olivier JH, Keane RW, Dietrich WD, Vaccari JPDR, Prasad A. Effects of iron accumulation and its chelation on oxidative stress in intracortical implants. Acta Biomater 2025:S1742-7061(25)00349-6. [PMID: 40355018 DOI: 10.1016/j.actbio.2025.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Long-term reliability of microelectrodes implanted in the cortex is hindered due to the foreign body response that occurs at the electrode-tissue interface. Following implantation, there is disruption of the blood-brain-barrier and vasculature, resulting in activation of immune cells and release of erythrocytes. As a result of hemolysis, erythrocytes degrade to heme and then to free iron. Excess free iron can participate in the Fenton Reaction, producing reactive oxygen species (ROS). Iron-mediated ROS production can contribute to oxidation of lipids, proteins, and DNA, facilitating a hostile environment of oxidative stress leading to oxidative cellular damage, cytotoxicity, and cell death. The objective of this study was to show the iron accumulation and the downstream effects of oxidative stress at the injury site. A 16-channel microelectrode array (MEA) was implanted in the rat somatosensory cortex. Our results indicated significant elevation of NOX complex subunits across timepoints, suggesting sustained oxidative stress. In a separate group of animals, we administered an iron chelator, deferoxamine mesylate (DFX), to evaluate the effects of chelation on iron accumulation, oxidative stress and damage, and neuronal survival. Results indicate that animals with iron chelation showed reduced ferric iron and markers of oxidative stress and damage corresponding with increased expression of neuronal cell bodies and electrophysiological functional performance. In summary, the study reveals the role of iron in mediating oxidative stress and the effects of modulating iron levels using iron chelation at the electrode-tissue interface. STATEMENT OF SIGNIFICANCE: Iron accumulation has been observed in central nervous system injuries and in neurodegenerative diseases such as Alzheimer's and Parkinson's disease. While the role of iron is studied in various neurodegenerative diseases and traumatic brain injury, iron accumulation and its effect on oxidative stress is not known for intracortical implants where there is a persistent injury due to the presence of a foreign device in the brain tissue. The study seeks to understand the effects of iron accumulation on oxidative stress and damage at the electrode-tissue interface in intracortical implants by using iron chelation as a method of modulating iron levels at the interface.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jordan L Grant
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Grant M Lee
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Serene Mattis
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Nicolas Gallardo
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Robert W Keane
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
4
|
Liu CP, Zheng S, Zhang P, Chen GH, Zhang YY, Sun HL, Peng L. Decreased serum SLC7A11 and GPX4 levels may reflect disease severity of acute ischaemic stroke. Ann Clin Biochem 2025; 62:191-201. [PMID: 39632577 DOI: 10.1177/00045632241305927] [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: 12/07/2024]
Abstract
ObjectiveThis study aimed to examine the levels of solute carrier family seven number 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) in the serum of patients with acute ischaemic stroke (AIS) and their relationship with disease severity.MethodsA total of 148 patients with AIS together with 148 healthy controls (HCs) were enrolled. The expression levels of SLC7A11 and GPX4 in serum were detected immediately as early as possible. Radiographic severity was detected by Alberta Stroke Program Early CT Score (ASPECTS). Disease severity was evaluated using modified Rankin Scale (mRS). High-sensitivity C-reactive protein (hs-CRP) and matrix metalloproteinase-9 (MMP-9) expression levels were also measured. A correlation analysis was conducted to determine the relationship between the expression levels of SLC7A11 and GPX4 with the clinical severity of the disease and the levels of hs-CRP and MMP-9. Furthermore, receiver operating characteristic (ROC) curve analysis was utilized to assess the potential of SLC7A11 and GPX4 as diagnostic markers.ResultsCompared to the HC group, the serum expression levels of SLC7A11 and GPX4 were significantly lower in the AIS group. Serum SLC7A11 levels were positively associated with serum GPX4 levels. The AIS group included 50 patients with mild neurological impairment, 52 with moderate neurological impairment, and 46 with severe neurological impairment. AIS patients with mild neurological impairment had drastically higher serum SLC7A11 and GPX4 levels compared with those with moderate neurological impairment. AIS patients with moderate neurological impairment showed significantly higher serum SLC7A11 and GPX4 concentrations compared with those with severe neurological impairment. ROC curve analysis demonstrated that both serum SLC7A11 and GPX4 may both act as potential indicators for evaluating of AIS disease severity. In addition, both serum SLC7A11 and GPX4 levels were positively correlated with ASPECTS. Both serum SLC7A11 and GPX4 levels were negatively associated with hs-CRP as well as MMP-9 levels. Serum SLC7A11 and GPX4 levels were significantly increased following comprehensive therapy.ConclusionsDecreased SLC7A11 and GPX4 levels may reflect disease severity of AIS.
Collapse
Affiliation(s)
| | - Su Zheng
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ping Zhang
- Department of Acupuncture, Shiyan Hospital of Traditional Chinese Medicine, Shiyan, China
| | - Guang-Hui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuan-Yuan Zhang
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Hui-Lin Sun
- Department of Radiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Peng
- Shiyan Hospital of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Shiyan, China
| |
Collapse
|
5
|
Gong F, Wei Y. LncRNA PVT1 promotes neuroinflammation after intracerebral hemorrhage by regulating the miR-128-3p/TXNIP axis. Int J Neurosci 2025; 135:573-587. [PMID: 38294729 DOI: 10.1080/00207454.2024.2312998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVE Intracerebral hemorrhage (ICH) has significant morbidity and mortality. TXNIP and the competing endogenous RNA (ceRNA) regulatory mechanism involved in long non-coding RNA (lncRNA) play roles in ICH. We probed the upstream microRNAs (miRNAs)/lncRNAs that regulated TXNIP expression in the ceRNA mechanism. METHODS ICH mouse model was established, and ICH secondary injury was simulated in BV2 microglia by hemin treatment. TXNIP was silenced 48 h before ICH modeling. The ICH mouse brain water content (BWC) and brain lesion volume after ICH were recorded. Neuronal apoptosis and neurological deficits were evaluated by double staining of NeuN and TUNEL/modified Garcia/corner turn/forelimb placement tests. Iba1 + microglia number and tumor necrosis factor-α (TNF-α)/interleukin-1β (IL-1β)/IL-10/TXNIP/PVT1/miR-128-3p levels were assessed by immunohistochemistry, Western blot, ELISA, and RT-qPCR. Cell viability/death of BV2 cells conditioned medium-treated neuron HT22 cells were assessed by CCK-8/LDH assays. miRNA that had a targeted binding relationship with TXNIP was screened. The targeted bindings of miR-128-3p to TXNIP/PVT1 to miR-128-3p were verified by dual-luciferase reporter gene assay. RESULTS TXNIP knockdown reduced post-ICH microglial activation/release of pro-inflammatory factors/brain edema/brain lesion volume/neurological deficits in mice and increased releases of anti-inflammatory factors. TXNIP/PVT1 knockdown inhibited hemin-induced inflammatory responses in BV2 cells and protected in vitro co-cultured HT22 cells. PVT1 was a sponge of miR-128-3p to repress TXNIP expression. miR-128-3p knockdown diminished PVT1 knockdown-inhibited hemin-induced BV2 cell inflammatory responses/neurotoxicity. CONCLUSIONS PVT1 silencing reduced hemin-induced neuroinflammation and had a protective effect on neurons by increasing the targeted inhibition of TXNIP by miR-128-3p.
Collapse
Affiliation(s)
- Fanyong Gong
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yiting Wei
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
6
|
Gong Y, Yang F, Liu Y, Gong Y. Ferroptosis-associated pathological injury mechanisms and therapeutic strategies after intracerebral hemorrhage. Front Neurol 2025; 16:1508718. [PMID: 40376154 PMCID: PMC12078154 DOI: 10.3389/fneur.2025.1508718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is an important neurological disease caused by the rupture of blood vessels in the brain parenchyma, with a high mortality and disability rate. At present, many studies have focused on the injury mechanisms and intervention strategies after ICH. However, there is no effective clinical treatment that can significantly improve the prognosis of ICH patients. Ferroptosis, a regulated form of cell death, has been identified as a significant contributor to brain tissues damage and neurological dysfunction following ICH. The hallmark of ferroptosis is iron-dependent lipid peroxidation, which is closely related to the pathological process of iron overload and oxidative stress after ICH. Exploring the interaction between ferroptosis and pathological injury mechanisms post-ICH will contribute to our understanding the key pathways involved in the ferroptosis-related injury mechanisms and facilitating the discovery of appropriate intervention strategies. On this basis, we present a comprehensive overview of ferroptosis-related brain injury mechanisms (e.g., iron overload, oxidative stress, inflammatory response and mass effect) in the pathogenesis and development of ICH. Following ICH, the degradation of hematoma and iron metabolism provide the fundamental material basis for ferroptosis, and oxidative stress primarily participates in the lipid peroxidation process of ferroptosis via related molecular pathways (such as the GPX4). By synthesizing current evidence, this article aims to provide a theoretical foundation for future research on therapeutic strategies targeting ferroptosis and related pathways in ICH.
Collapse
Affiliation(s)
- Yuhua Gong
- School of Smart Health, Chongqing Polytechnic University of Electronic Technology, Chongqing, China
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fumei Yang
- School of Smart Health, Chongqing Polytechnic University of Electronic Technology, Chongqing, China
| | - Ying Liu
- School of Smart Health, Chongqing Polytechnic University of Electronic Technology, Chongqing, China
| | - Yuping Gong
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Schüssler SC, Paul A, Niederreiter U, Deiters L, Fahlbusch FB, Morhart P, Trollmann R. Seizures in preterm infants with germinal-matrix-intraventricular hemorrhage (GM-IVH): a retrospective monocentric study on predictors and neurodevelopmental outcome. Eur J Paediatr Neurol 2025; 56:51-57. [PMID: 40311512 DOI: 10.1016/j.ejpn.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/03/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
AIM Germinal-matrix-intraventricular hemorrhage (GM-IVH) is a leading cause of seizures in preterm infants. This study aimed to analyze risk factors associated with seizures and to evaluate neurodevelopmental outcomes in preterm infants with GM-IVH and seizures. METHODS We conducted a retrospective study from 2011 to 2019, identifying preterm infants with GM-IVH grades 2-4 through an electronic patient file system. Seizures were diagnosed based on clinical manifestations and abnormal EEG findings. Infants were grouped by the presence or absence of seizures, and associated comorbidities were compared. Neurodevelopmental follow-up was assessed at two years of age using the Mental Bayley Scales of Infant Development II (BSID-II). Outcomes of infants with seizures were compared to all tested preterm infants with birth weight <1500 g born between 2011 and 2019 (n = 195). RESULTS A total of 34 preterm infants with GM-IVH grades 2-4 were included. Seizures occurred in 52.9 % of cases. Their occurrence was significantly associated with lower gestational age (mean 28.1 vs. 30 weeks, p = 0.04) and pneumonia (p = 0.003). Infants with seizures had significantly lower BSID-II Mental scores (n = 15) compared to those without seizures (86.3 ± 18.3 vs. 104.9 ± 8.5, p = 0.03). However, as these infants had a lower gestational age, we could not distinguish if they had a poorer outcome because of seizures or because of immaturity. CONCLUSION Seizures in preterm infants with GM-IVH were significantly associated with lower gestational age and pneumonia. Infections and inflammation may contribute to seizure development. Larger studies with continuous EEG monitoring are needed to validate these findings.
Collapse
Affiliation(s)
- Stephanie C Schüssler
- Department of Pediatrics and Adolescent Medicine, Pediatric Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - Anna Paul
- Department of Pediatrics and Adolescent Medicine, Pediatric Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - Undine Niederreiter
- Department of Pediatrics and Adolescent Medicine, Pediatric Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - Ludger Deiters
- Department of Pediatrics and Adolescent Medicine, Pediatric Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - Fabian B Fahlbusch
- Neonatology and Pediatric Intensive Care, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany; Department of Pediatrics and Adolescent Medicine, Neonatology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - Patrick Morhart
- Department of Pediatrics and Adolescent Medicine, Neonatology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - Regina Trollmann
- Department of Pediatrics and Adolescent Medicine, Pediatric Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| |
Collapse
|
8
|
Jiang M, Cao H, Chen W, Yu Y, Lin J. Inhibition of ACSL4 Attenuates Behavioral Deficits by Regulating Ferroptosis in a Murine Model of Systemic Lupus Erythematosus. Int J Mol Sci 2025; 26:3553. [PMID: 40332005 PMCID: PMC12027207 DOI: 10.3390/ijms26083553] [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: 03/03/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is a disorder with a poor prognosis characterized by psychiatric and neurological manifestations directly associated with systemic lupus erythematosus (SLE). Neutrophil ferroptosis has been identified as a significant contributor to neutropenia and disease progression in SLE, but its role in NPSLE remains unclear. Female MRL/lpr and MRL/Mpj mice were used. The selective ferroptosis inhibitor liproxstatin-1 and the acyl-CoA synthetase long-chain family member 4 (ACSL4) inhibitor rosiglitazone were administered separately. Assessments included behavioral testing, transmission electron microscopy (TEM), ELISA, Western blotting, RT-PCR, and Nissl staining. Our data showed that neurons in the brain parenchyma undergo ferroptosis, with decreased glutathione peroxidase 4 (GPX4) expression and increased levels of lipid peroxidation indicators and have the typical morphology of ferroptosis confirmed by transmission electron microscopy. Selective ferroptosis inhibitor liproxstatin-1 attenuated the neuropsychiatric manifestations, including depression-like and impulsive behaviors, of MRL/lpr mice. ACSL4 is the main enzyme in lipid metabolism. Our study further found that the utilization of rosiglitazone by inhibiting ACSL4 could also significantly attenuate neuropsychiatric manifestations of MRL/lpr mice. Moreover, blocking ACSL4 might considerably boost GPX4 levels and decrease lipid peroxidation indicators in NPSLE, with reduced neuronal damage, as well as reduced neuroinflammation. This study concluded that inhibiting ACSL4 could facilitate the recuperation of behavioral deficits by suppression of ferroptosis in NPSLE, implying that ACSL4 might be a potential new therapeutic focus for NPSLE.
Collapse
Affiliation(s)
- Mengdi Jiang
- Department of Rheumatology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | | | | | - Jin Lin
- Department of Rheumatology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
9
|
Huang R, Pang Q, Zheng L, Lin J, Li H, Wan L, Wang T. Cholesterol metabolism: physiological versus pathological aspects in intracerebral hemorrhage. Neural Regen Res 2025; 20:1015-1030. [PMID: 38989934 PMCID: PMC11438341 DOI: 10.4103/nrr.nrr-d-23-01462] [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: 08/31/2023] [Revised: 12/19/2023] [Accepted: 01/27/2024] [Indexed: 07/12/2024] Open
Abstract
Cholesterol is an important component of plasma membranes and participates in many basic life functions, such as the maintenance of cell membrane stability, the synthesis of steroid hormones, and myelination. Cholesterol plays a key role in the establishment and maintenance of the central nervous system. The brain contains 20% of the whole body's cholesterol, 80% of which is located within myelin. A huge number of processes (e.g., the sterol regulatory element-binding protein pathway and liver X receptor pathway) participate in the regulation of cholesterol metabolism in the brain via mechanisms that include cholesterol biosynthesis, intracellular transport, and efflux. Certain brain injuries or diseases involving crosstalk among the processes above can affect normal cholesterol metabolism to induce detrimental consequences. Therefore, we hypothesized that cholesterol-related molecules and pathways can serve as therapeutic targets for central nervous system diseases. Intracerebral hemorrhage is the most severe hemorrhagic stroke subtype, with high mortality and morbidity. Historical cholesterol levels are associated with the risk of intracerebral hemorrhage. Moreover, secondary pathological changes after intracerebral hemorrhage are associated with cholesterol metabolism dysregulation, such as neuroinflammation, demyelination, and multiple types of programmed cell death. Intracellular cholesterol accumulation in the brain has been found after intracerebral hemorrhage. In this paper, we review normal cholesterol metabolism in the central nervous system, the mechanisms known to participate in the disturbance of cholesterol metabolism after intracerebral hemorrhage, and the links between cholesterol metabolism and cell death. We also review several possible and constructive therapeutic targets identified based on cholesterol metabolism to provide cholesterol-based perspectives and a reference for those interested in the treatment of intracerebral hemorrhage.
Collapse
Affiliation(s)
- Ruoyu Huang
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| | - Qiuyu Pang
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| | - Lexin Zheng
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiaxi Lin
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| | - Hanxi Li
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| | - Lingbo Wan
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| | - Tao Wang
- Department of Forensic Science, School of Basic Medicine and Biological Sciences, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
10
|
Zhuo B, Qin C, Deng S, Jiang H, Si S, Tao F, Cai F, Meng Z. The role of ACSL4 in stroke: mechanisms and potential therapeutic target. Mol Cell Biochem 2025; 480:2223-2246. [PMID: 39496916 PMCID: PMC11961533 DOI: 10.1007/s11010-024-05150-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/24/2024] [Indexed: 11/06/2024]
Abstract
Stroke, as a neurological disorder with a poor overall prognosis, has long plagued the patients. Current stroke therapy lacks effective treatments. Ferroptosis has emerged as a prominent subject of discourse across various maladies in recent years. As an emerging therapeutic target, notwithstanding its initial identification in tumor cells associated with brain diseases, it has lately been recognized as a pivotal factor in the pathological progression of stroke. Acyl-CoA synthetase long-chain family member 4 (ACSL4) is a potential target and biomarker of catalytic unsaturated fatty acids mediating ferroptosis in stroke. Specifically, the upregulation of ACSL4 leads to heightened accumulation of lipid peroxidation products and reactive oxygen species (ROS), thereby exacerbating the progression of ferroptosis in neuronal cells. ACSL4 is present in various tissues and involved in multiple pathways of ferroptosis. At present, the pharmacological mechanisms of targeting ACSL4 to inhibit ferroptosis have been found in many drugs, but the molecular mechanisms of targeting ACSL4 are still in the exploratory stage. This paper introduces the physiopathological mechanism of ACSL4 and the current status of the research involved in ferroptosis crosstalk and epigenetics, and summarizes the application status of ACSL4 in modern pharmacology research, and discusses the potential application value of ACSL4 in the field of stroke.
Collapse
Affiliation(s)
- Bifang Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chenyang Qin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shizhe Deng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hailun Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shangkun Si
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng Tao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fei Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhihong Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
11
|
Mao N, Zhang M, Shen M, Yuan J, Lin Z. Research progress on ferroptosis in cerebral hemorrhage. Biomed Pharmacother 2025; 185:117932. [PMID: 40015051 DOI: 10.1016/j.biopha.2025.117932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
The pathophysiology of intracerebral hemorrhage (ICH) is complex and can cause variable degrees of cell death. Recently, ferroptosis, an emerging cell death mechanism, has garnered significant attention in cerebral hemorrhage disorder. This study aimed to examine iron mortality after cerebral hemorrhage and current targets for potential therapeutic interventions. We specifically focused on iron metabolism abnormalities, lipid peroxidation, and related neuroinflammation and introduced molecular mechanisms, including transcription factors, to gain a better understanding of the underlying mechanisms of ferroptosis and investigate possible therapeutic options for ICH.
Collapse
Affiliation(s)
- Niping Mao
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Zhang
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Shen
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junhui Yuan
- Department of Neonatology, Wenling maternal and child health care hospital, Wenling, Zhejiang, China.
| | - Zhenlang Lin
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
12
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [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: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
An J, Liu Z, Wang Y, Meng K, Wang Y, Sun H, Li M, Tang Z. Drug delivery strategy of hemostatic drugs for intracerebral hemorrhage. J Control Release 2025; 379:202-220. [PMID: 39793654 DOI: 10.1016/j.jconrel.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Intracerebral hemorrhage (ICH) is associated with high rates of mortality and disability, underscoring an urgent need for effective therapeutic interventions. The clinical prognosis of ICH remains limited, primarily due to the absence of targeted, precise therapeutic options. Advances in novel drug delivery platforms, including nanotechnology, gel-based systems, and exosome-mediated therapies, have shown potential in enhancing ICH management. This review delves into the pathophysiological mechanisms of ICH and provides a thorough analysis of existing treatment strategies, with an emphasis on innovative drug delivery approaches designed to address critical pathological pathways. We assess the benefits and limitations of these therapies, offering insights into future directions in ICH research and highlighting the transformative potential of next-generation drug delivery systems in improving patient outcomes.
Collapse
Affiliation(s)
- Junyan An
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhilin Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yihan Wang
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China
| | - Ke Meng
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China
| | - Yixuan Wang
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China
| | - Hai Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Miao Li
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
14
|
Mouli K, Liopo AV, McHugh EA, Underwood E, Zhao J, Dash PK, Vo ATT, Malojirao VH, Hegde ML, Tour JM, Derry PJ, Kent TA. Oxidized Carbon Nanoparticles Enhance Cellular Energetics With Application to Injured Brain. Adv Healthc Mater 2025; 14:e2401629. [PMID: 39329414 PMCID: PMC11937864 DOI: 10.1002/adhm.202401629] [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: 05/02/2024] [Revised: 07/26/2024] [Indexed: 09/28/2024]
Abstract
Pro-energetic effects of functionalized, oxidized carbon nanozymes (OCNs) are reported. OCNs, derived from harsh acid oxidation of single-wall carbon nanotubes or activated charcoal are previously shown to possess multiple nanozymatic activities including mimicking superoxide dismutase and catalyzing the oxidation of reduced nicotinamide adenine dinucleotide (NADH) to NAD+. These actions are predicted to generate a glycolytic shift and enhance mitochondrial energetics under impaired conditions. Impaired mitochondrial energy metabolism is increasingly recognized as an important facet of traumatic brain injury (TBI) pathophysiology and decreases the efficiency of electron transport chain (ETC)-coupled adenosine triphosphate (ATP) and NAD+ regeneration. In vitro, OCNs promote a pro-aerobic shift in energy metabolism that persists through ETC inhibition and enhances glycolytic flux, glycolytic ATP production, and cellular generation of lactate, a crucial auxiliary substrate for energy metabolism. To address specific mechanisms of iron injury from hemorrhage, OCNs with the iron chelator, deferoxamine (DEF), covalently-linked were synthesized. DEF-linked OCNs induce a glycolytic shift in-vitro and in-vivo in tissue sections from a rat model of TBI complicated by hemorrhagic contusion. OCNs further reduced hemorrhage volumes 3 days following TBI. These results suggest OCNs are promising as pleiotropic mediators of cell and tissue resilience to injury.
Collapse
Affiliation(s)
- Karthik Mouli
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
| | - Anton V. Liopo
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- Department of ChemistryRice UniversityHoustonTX77005USA
| | - Emily A. McHugh
- Department of ChemistryRice UniversityHoustonTX77005USA
- Smalley‐Curl InstituteRice UniversityHoustonTX77005USA
| | - Erica Underwood
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Jing Zhao
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Pramod K. Dash
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Anh T. T. Vo
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
| | - Vikas H. Malojirao
- Center for NeuroregenerationDepartment of NeurosurgeryDivision of DNA Repair ResearchHouston Methodist Research InstituteHoustonTX77030USA
| | - Muralidhar L. Hegde
- Center for NeuroregenerationDepartment of NeurosurgeryDivision of DNA Repair ResearchHouston Methodist Research InstituteHoustonTX77030USA
- Department of NeurosciencesWeill Cornell Medical CollegeNew YorkNYUSA
- EnMedSchool of Engineering MedicineTexas A&M UniversityHouston77030USA
| | - James M. Tour
- Department of ChemistryRice UniversityHoustonTX77005USA
- Smalley‐Curl InstituteRice UniversityHoustonTX77005USA
- Welch Institute for Advanced MaterialsRice UniversityHoustonTX77005USA
- The NanoCarbon CenterRice UniversityHoustonTX77005USA
| | - Paul J. Derry
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- EnMedSchool of Engineering MedicineTexas A&M UniversityHouston77030USA
| | - Thomas A. Kent
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- Department of ChemistryRice UniversityHoustonTX77005USA
- Stanley H. Appel Department of NeurologyHouston Methodist Hospital and Research InstituteHoustonTX77030USA
| |
Collapse
|
15
|
Liu X, Chen G, Yang Y, Liu F, Wu G, An L, Tang T, Zhang J. Comprehensive multi-omics analysis reveals the mechanism of hepatotoxicity induced by Emilia sonchifolia (L.) DC. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119371. [PMID: 39826791 DOI: 10.1016/j.jep.2025.119371] [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: 04/26/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Emilia sonchifolia is a very widely used traditional Chinese medicine, with the efficacy of heat-clearing, detoxicating, dissipating blood stasis, reducing swelling and relieving pain. As a widely used traditional miao herb, Emilia sonchifolia is often used to treat upper respiratory tract infections, oral ulcer, pneumonia, mastitis, enteritis, bacillum, urinary tract infection, sores, eczema, falls and injuries, etc. In fact, many cases of liver injury caused by Emilia sonchifolia have been reported clinically. However, the mechanisms underlying hepatotoxicity induced by Emilia sonchifolia remain poorly understood. AIM OF THE STUDY This study aimed to systematically evaluate the acute and chronic hepatotoxicity of water extract from Emilia sonchifolia, identify its hepatotoxic metabolites, and elucidate the potential mechanisms underlying Emilia sonchifolia-induced hepatotoxicity. MATERIAL AND METHOD The chemical components in the water extract of Emilia sonchifolia were identified using mass spectrometry. The acute toxicity study was conducted by orally administering a gradient dose of water extract of Emilia sonchifolia ranging from 0 to 37.6 g/kg. Mice were orally administered a water extract of Emilia sonchifolia at a dose of 13.72 g/kg/d for 14 days to induce liver injury. The hepatotoxicity was evaluated using hematoxylin and eosin staining as well as enzyme-linked immunosorbent assay (ELISA). The mechanisms of hepatotoxicity were explored through transcriptomics, proteomics, and metabolomics analysis. Meanwhile, the core pathways related to the hepatotoxicity of Emilia sonchifolia were analyzed and validated using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and ELISA. RESULT The present study demonstrates that the water extract of Emilia sonchifolia can induce hepatotoxicity in mice. We found that the water extract of Emilia sonchifolia contained hepatotoxic pyrrolizidine alkaloids, such as seneciphyllin, senecionine, rinderine, echimidine, retrorsine and echimidine N-oxide. A dose of 19.20 g/kg or higher of the water extract of Emilia sonchifolia caused acute liver failure and death in mice. A dose of 13.72 g/kg or lower of the water extract of Emilia sonchifolia produced dose-dependent acute hepatotoxicity. Meanwhile, a dose of 13.72 g/kg of the water extract from Emilia sonchifolia induced chronic hepatotoxicity in mice. Furthermore, the results of liver transcriptomics, proteomics, and metabolomics indicate that the mechanism of hepatotoxicity induced by the water extract of Emilia sonchifolia is associated with ferroptosis caused by abnormalities in bile acid accumulation, lipid and bilirubin accumulation, and glutathione metabolism. The validation experiment results demonstrate that in mice treated with the water extract of Emilia sonchifolia, the gene levels of Cyp2c29, Cyp3a41a and Ugt2b1 decreased while the gene level of Hsd3b3 increased. In mice treated with a water extract of Emilia sonchifolia, the levels of total bilirubin, direct bilirubin, total bile acids, alkaline phosphatase, and γ-glutamyl transferase were significantly elevated. Additionally, in mice treated with a water extract of Emilia sonchifolia, the levels of malondialdehyde increased while the levels of catalase and superoxide dismutase decreased. CONCLUSION In conclusion, our results suggest that the water extract of Emilia sonchifolia can cause hepatotoxicity in mice. The chronic hepatotoxicity of Emilia sonchifolia is associated with Cyp2c29, Cyp3a41a, Ugt2b1, and Hsd3b3-mediated cholestasis, oxidative stress, and ferroptosis.
Collapse
Affiliation(s)
- Xin Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Gongzhen Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Yuqi Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Feng Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Guangzhou Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Lili An
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Ting Tang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
16
|
Liu Q, Han Z, Li T, Meng J, Zhu C, Wang J, Wang J, Zhang Z, Wu H. Microglial HO-1 aggravates neuronal ferroptosis via regulating iron metabolism and inflammation in the early stage after intracerebral hemorrhage. Int Immunopharmacol 2025; 147:113942. [PMID: 39740507 DOI: 10.1016/j.intimp.2024.113942] [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: 06/04/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Heme oxygenase 1 (HO-1), an enzyme involved in heme catabolism, has been shown upregulated in microglia cells and plays a critical roles in neurological damages after intracerebral hemorrhage (ICH). However, the mechanisms by which HO-1 mediates the neuronal damages are still obscure. Here, our findings demonstrate that HO-1 over-expression exacerbates the pro-inflammatory response of microglia and induces neuronal ferroptosis through promoting intracellular iron deposition in the ICH model both in vitro and in vivo. Furthermore, in the co-cultured ICH model in vitro, we verify that HO-1 over-expression disrupts the balance of iron metabolism in microglia, which increases the iron efflux to the extracellular space and promotes iron ion uptake in neurons, leading to lipid peroxidation injury and further contributing to neuronal ferroptosis. Moreover, the specific ferroptosis inhibitor Ferrostatin-1 (Fer-1) treatment could mitigate the damages in the co-cultured HT22 cells that caused by HO-1 over-expression in microglia, and improve the neurological function in the ICH model in mice. By shedding light on the mechanisms of aggravating neuronal ferroptosis due to HO-1 overexpression in the early stages after ICH, our study provides insights into the potential therapy of targeting HO-1 to treat ICH.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ziyi Han
- College of Medical Laboratory Technology, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tao Li
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jincheng Meng
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chenwei Zhu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Zhen Zhang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - He Wu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
17
|
Zeng Y, Liu J, Kong Z, Han G, Xiong Y, Luo T, Chu L, Zhang P, Ma D, Lan J, Liu G, Liu J, Zhang J, Tan Y. Catechin-Based Polyphenol Nanoparticles Ameliorated Ferroptosis to Alleviate Brain Injury after Intracerebral Hemorrhage. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7424-7437. [PMID: 39849318 DOI: 10.1021/acsami.4c19513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Spontaneous intracerebral hemorrhagic stroke (ICH) is a highly aggressive disease, with a high incidence and mortality rate. Iron deposition following ICH leads to oxidative damage and motor dysfunction, significantly impacting the overall quality of life for those affected. Here, a polyphenolic nanomedicine, catechin-based polyphenol nanoparticles surface-modified by thiol-terminated poly(ethylene glycol) (CNPs@PEG), was developed through the oxidative polymerization and self-assembly of catechin, a natural compound in tea. Due to its potent antioxidant and metal-chelating properties, CNPs@PEG effectively maintained blood-brain barrier integrity, reduced brain edema, significantly increased the survival rate of mice with cerebral hemorrhage and markedly improved neurological deficits after ICH. Mechanistically, CNPs@PEG accomplishes this by chelating iron, enhancing tissue antioxidant capacity, reducing oxidative stress, and inhibiting iron deposition. This approach holds promise as a targeted therapeutic strategy for addressing cerebral hemorrhage and other conditions associated with iron overload.
Collapse
Affiliation(s)
- Yu Zeng
- Medical College of Guizhou University, Guiyang 550000, China
| | - Jian Liu
- Graduate School of Zunyi Medical University, Zunyi 563000, China
| | - Zhuo Kong
- Department of Surgery, Clinical Medical College, Guizhou Medical University, Guiyang 550000, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Yunbiao Xiong
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Tao Luo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Liangzhao Chu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Peng Zhang
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Dongxu Ma
- Department of Neurosurgery, Sansui People's Hospital, Qiandongnan Miao and Dong Autonomous Prefecture 556000, China
| | - Jinhai Lan
- Department of the Second Surgery, Ziyun People's Hospital, Anshun 550800, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Jian Liu
- Medical College of Guizhou University, Guiyang 550000, China
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| |
Collapse
|
18
|
Fan X, Liu S, Yu J, Hua J, Feng Y, Wang Z, Shen Y, Lan W, Wang J. Puerarin Ameliorates the Ferroptosis in Diabetic Liver Injure Through the JAK2/STAT3 Pathway Inhibition Based on Network Pharmacology and Experimental Validation. Drug Des Devel Ther 2025; 19:737-757. [PMID: 39911447 PMCID: PMC11796443 DOI: 10.2147/dddt.s487496] [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: 08/19/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
Background Diabetic liver injury (DLI) is a common complication of diabetes mellitus (DM), which seriously endangers the health of diabetic patients. Puerarin, the main active component of Pueraria lobata, has shown positive effects in lowering blood glucose and lipids, resisting oxidative stress, and protecting the liver. However, the mechanism of protective effect of Puerarin on DLI remains unclear. Methods Various databases were used to screen for targets of Puerarin, ferroptosis and DLI. Protein-protein interaction (PPI) network and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used to predict key targets and pathways. Molecular docking was used to predict the interactions between Puerarin and core targets. KK/Upj-Ay/J (KKAy) mice and high glucose (HG)-induced AML12 cells were used to study the protective effect of Puerarin on DLI. The molecular mechanisms by which Puerarin acts were further verified by in vivo and in vitro experiments. Results KEGG analysis indicated that the JAK/STAT pathway might be related to the anti-DLI effect of Puerarin. Molecular docking revealed that Puerarin has good affinity for JAK2 and STAT3. In vivo, Puerarin (80 mg/kg) reduced body weight, blood glucose, blood lipids and liver function in KKAy mice fed a high-sugar, high-fat diet. Puerarin also ameliorated hepatic pathological changes and inflammatory responses, and attenuated oxidative stress and iron overload in KKAy mice. Western blotting results showed that Puerarin could regulate the expression of proteins related to JAK2/STAT3 pathway and ferroptosis pathway. In vitro, Puerarin (25, 50, 100 μM) increased cell viability and decreased steatosis and liver function indexes in AML12 cells induced by HG (30 mm) to varying degrees. More importantly, AG490 blocker experiments showed that the regulation of ferroptosis process by Puerarin was dependent on the JAK2/STAT3 pathway. Conclusion In conclusion, this study revealed Puerarin may regulate the ferroptosis process by inhibiting the JAK2/STAT3 pathway for the treatment of DLI.
Collapse
Affiliation(s)
- Xiaoxu Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Shuangqiao Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jing Yu
- School of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Jian Hua
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yingtong Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Zhen Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yiwei Shen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wei Lan
- School of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Jingxia Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
19
|
Chen J, Wang L, Peng X, Cheng T, Yang Y, Su J, Zou H, Wang S, Mao Y, Wu L, Yin X, Li M, Zhu M, Zhou W. Identification of CSPG4 as a Biomarker and Therapeutic Target for Infantile Post-Hemorrhagic Hydrocephalus via Multi-Omics Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410056. [PMID: 39686677 PMCID: PMC11809374 DOI: 10.1002/advs.202410056] [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] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Intraventricular hemorrhage in preterm neonates has become a major global health problem and is associated with a high risk of post-hemorrhagic hydrocephalus (PHH). Identifying diagnostic markers and therapeutic targets is a focal challenge in the PHH prevention and control. Here, this study applies multi-omics analyses to characterize the biochemical, proteomic, and metabolomic profiles of the cerebrospinal fluid (CSF) in clinical human cohorts to investigate disease development and recovery processes occurring due to PHH. Integrative multiomics analysis suggests that the over-representation of ferroptosis, calcium, calcium ion binding, and cell adhesion signaling pathways is associated with PHH. Bioinformatic analysis indicates that chondroitin sulfate proteoglycan 4 (CSPG4) is discovered as a CSF biomarker and positively correlated with the ventricular size and the rate of periventricular leukomalacia. Next, it is further demonstrated that these signaling pathways are dysregulated in the choroid plexus (ChP) in PHH by using in vitro cellular experiments and rat models of PHH, whereas CSPG4 silencing can suppress ferroptosis, cell adhesion function, and the intracellular flow of Ca2+. These findings broaden the understanding of the pathophysiological mechanisms of PHH and suggest that CSPG4 may be an effective therapeutic target for PHH.
Collapse
Affiliation(s)
- Juncao Chen
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Lin Wang
- Department of RadiologyGuangzhou Women and Children's Medical CenterGuangdong Provincial Clinical Research Center for Child HealthGuangzhou510623China
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Xiangwen Peng
- Changsha Hospital for Maternal and Child HealthcareChangsha410100China
| | - Tingting Cheng
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Yihui Yang
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Jingzhen Su
- Department of NeonatologyDongguan Maternal and Child Health HospitalDongguan523057China
| | - Hongmei Zou
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Siyao Wang
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Yueting Mao
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Lixiang Wu
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Xuntao Yin
- Department of RadiologyGuangzhou Women and Children's Medical CenterGuangdong Provincial Clinical Research Center for Child HealthGuangzhou510623China
| | - Minxu Li
- Department of NeonatologyDongguan Maternal and Child Health HospitalDongguan523057China
| | - Mingwei Zhu
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
- Key Laboratory of Developmental Disorders in ChildrenLiuzhou Maternity and Child Healthcare HospitalLiuzhou545006China
| | - Wei Zhou
- Department of NeonatologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| |
Collapse
|
20
|
Zhou Y, Li J, Yuan Y, Zhang H, Luo X, Wang F, Tao Y, Yue J, Huang L, Wu L, Cao Y, Yu Q, He Q. Metrnl/C-KIT Axis Attenuates Early Brain Injury Following Subarachnoid Hemorrhage by Inhibiting Neuronal Ferroptosis. CNS Neurosci Ther 2025; 31:e70286. [PMID: 39981761 PMCID: PMC11843251 DOI: 10.1111/cns.70286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Ferroptosis is a distinct form of cell death characterized by iron-dependent lipid peroxidation and plays a crucial role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH). As a newly discovered endogenous ligand for the C-KIT receptor tyrosine kinase, meteorin-like protein (Metrnl) exerts regulatory functions in oxidative stress and protects against various diseases. However, the specific role of the Metrnl/C-KIT axis in neuronal ferroptosis during EBI following SAH remains to be elucidated. METHODS Sprague Dawley rats were used to establish the SAH model through endovascular perforation. r-Metrnl was administered intranasally 1 h after SAH. Metrnl shRNA, C-KIT inhibitor ISCK03, AMPK inhibitor dorsomorphin, and Nrf2 inhibitor ML385 were administered intracerebroventricularly or intraperitoneally before r-Metrnl treatment to explore the underlying mechanisms. Neurobehavioral assessments, immunofluorescence, western blot, ELISA, Fluoro-Jade C staining, transmission electron microscopy, and Nissl staining were conducted to evaluate the effects. Additionally, primary neuron culture with hemoglobin (Hb) stimulation was used for in vitro studies. RESULTS Phosphorylated C-KIT and endogenous Metrnl levels were upregulated after SAH. Knockdown of Metrnl aggravated neurobehavioral deficits and neuronal ferroptosis, whereas r-Metrnl treatment showed a protective effect. Mechanistically, r-Metrnl significantly increased the protein levels of SLC7A11, GPX4, FTH, FSP1, and GSH, whereas it decreased the levels of ACSL4, 4HNE, and MDA in the ipsilateral hemisphere 24 h after SAH. Also, r-Metrnl reduced mitochondrial shrinkage, increased mitochondrial crista, and decreased membrane density. However, the beneficial effects of r-Metrnl were partially reversed by ISCK03, dorsomorphin, or ML385 treatment both in vivo and in vitro. CONCLUSIONS Our study demonstrated that r-Metrnl reduced neuronal ferroptosis and improved neurological outcomes after SAH by modulating the C-KIT/AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- You Zhou
- Department of Critical Care Medicine, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Jiani Li
- Department of Neurology, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Ye Yuan
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Xu Luo
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Feng Wang
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Jianhe Yue
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Luyi Huang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Lei Wu
- Department of NeurologyGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
| | - Yunxing Cao
- Department of Critical Care Medicine, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Qian Yu
- Department of Neurosurgery, School of Medicine, The Second Affiliated HospitalZhejiang UniversityHangzhouZhejiangChina
| | - Qiuguang He
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| |
Collapse
|
21
|
Chiang PT, Tsai LK, Tsai HH. New targets in spontaneous intracerebral hemorrhage. Curr Opin Neurol 2025; 38:10-17. [PMID: 39325041 PMCID: PMC11706352 DOI: 10.1097/wco.0000000000001325] [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: 09/27/2024]
Abstract
PURPOSE OF REVIEW Intracerebral hemorrhage (ICH) is a devastating stroke with limited medical treatments; thus, timely exploration of emerging therapeutic targets is essential. This review focuses on the latest strategies to mitigate secondary brain injury post-ICH other than targeting surgery or hemostasis, addressing a significant gap in clinical practice and highlighting potential improvements in patient outcomes. RECENT FINDINGS Promising therapeutic targets to reduce secondary brain injury following ICH have recently been identified, including attenuation of iron toxicity and inhibition of ferroptosis, enhancement of endogenous resorption of hematoma, and modulation of perihematomal inflammatory responses and edema. Additionally, novel insights suggest the lymphatic system of the brain may potentially play a role in hematoma clearance and edema management. Various experimental and early-phase clinical trials have demonstrated these approaches may potentially offer clinical benefits, though most research remains in the preliminary stages. SUMMARY Continued research is essential to identify multifaceted treatment strategies for ICH. Clinical translation of these emerging targets could significantly enhance the efficacy of therapeutic interventions and potentially reduce secondary brain damage and improve neurological recovery. Future efforts should focus on large-scale clinical trials to validate these approaches, to pave the way for more effective treatment protocols for spontaneous ICH.
Collapse
Affiliation(s)
- Pu-Tien Chiang
- Department of Neurology, National Taiwan University Hospital
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital
| | - Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital
| |
Collapse
|
22
|
Liu L, Jia P, Liu T, Liang J, Dang Y, Rastegar-Kashkooli Y, Li Q, Liu J, Man J, Zhao T, Xing N, Wang F, Chen X, Zhang J, Jiang C, Zille M, Zhang Z, Fan X, Wang J, Wang J. Metabolic dysfunction contributes to mood disorders after traumatic brain injury. Ageing Res Rev 2025; 104:102652. [PMID: 39746403 DOI: 10.1016/j.arr.2024.102652] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Traumatic brain injury (TBI) presents significant risks concerning mortality and morbidity. Individuals who suffer from TBI may exhibit mood disorders, including anxiety and depression. Both preclinical and clinical research have established correlations between TBI and disturbances in the metabolism of amino acids, lipids, iron, zinc, and copper, which are implicated in the emergence of mood disorders post-TBI. The purpose of this review is to elucidate the impact of metabolic dysfunction on mood disorders following TBI and to explore potential strategies for mitigating anxiety and depression symptoms. We researched the PubMed and Web of Science databases to delineate the mechanisms by which metabolic dysfunction contributes to mood disorders in the context of TBI. Particular emphasis was placed on the roles of glutamate, kynurenine, lipids, iron, zinc, and copper metabolism. Metabolic dysfunction is linked to mood disorders post-TBI through multiple pathways, encompassing the glutamatergic system, the kynurenine pathway, endocannabinoids, iron deposition, iron-related ferroptosis, zinc deficiency, and copper dysregulation. Furthermore, this review addresses the influence of metabolic dysfunction on mood disorders in the elderly demographic following TBI. Targeting metabolic dysfunction for therapeutic intervention appears promising in alleviating symptoms of anxiety and depression that arise after TBI. While further investigation is warranted to delineate the underlying pathophysiologic mechanisms of mood disorders post-TBI, current evidence underscores the potential contribution of metabolic dysfunction to these conditions. Therefore, rectifying metabolic dysfunction represents a viable and strategic approach to addressing mood disorders following TBI.
Collapse
Affiliation(s)
- Lang Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Tongzhou Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiaxin Liang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yijia Dang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China.
| | - Jingqi Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiang Man
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Ting Zhao
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan 610060, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiewen Zhang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Chao Jiang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria.
| | - Zhenhua Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
23
|
Hu N, Zhang G, An L, Wang W, An R, Li Y. PF4 inhibits ferroptosis-mediated intracerebral hemorrhage through modulating the CXCR3/AKT1/SLC7A11 signaling pathway. BIOMOLECULES & BIOMEDICINE 2025; 25:563-577. [PMID: 39558855 PMCID: PMC12010983 DOI: 10.17305/bb.2024.11283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis plays a crucial role in the secondary pathophysiological damage to brain tissue surrounding hematomas after intracerebral hemorrhage (ICH). While platelet factor 4 (PF4) is known to promote regeneration following peripheral nerve injury, its role in brain tissue repair after cerebral hemorrhage remains unclear. In this study, Hemin-induced PC12 cells were treated with various inhibitors and assessed for viability, oxidative stress, and ferroptosis using a combination of assays, including CCK-8 (Cell Counting Kit-8), EdU (5-Ethynyl-2'-deoxyuridine), flow cytometry, and immunofluorescence. ICH cells were also treated with recombinant PF4 (Rm-PF4) and a CXCR3 antagonist (AMG487) to investigate the mechanism by which Rm-PF4 influences Hemin-induced PC12 cell injury and inflammation. Subsequently, ICH mouse models were established via collagenase injection. Neurological function in these mice was evaluated using the Cylinder and Corner tests. Histopathological damage to brain tissue was analyzed through HE, TUNEL, and Nissl staining, as well as immunohistochemistry, to further explore the role of Rm-PF4 in controlling neuroinflammation in vivo. Results showed that Rm-PF4 inhibited Hemin-mediated ferroptosis-induced PC12 cell damage and inflammation by activating the CXCR3/AKT1/SLC7A11 signaling pathway. Blocking the CXCR3/AKT1/SLC7A11 pathway partially reversed PF4's protective effects on Hemin-induced PC12 cells.In ICH mice, pro-inflammatory marker CD16 (3rd day) and anti-inflammatory marker Arg1 (7th day) were significantly decreased and increased, respectively (p<0.05). IL-6, TNF-α, and IL-1β levels were down-regulated in brain tissues after Rm-PF4 injection, which was significantly reversed by AMG487. PF4 inhibits ferroptosis after ICH reduced PC12 cell damage and the inflammatory response via activating the CXCR3/AKT1/SLC7A11 pathway.
Collapse
Affiliation(s)
- Na Hu
- Department of Biochemistry and Biology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Guohong Zhang
- Department of Biochemistry and Biology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Liping An
- Department of Biochemistry and Biology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Wei Wang
- Department of Biochemistry and Biology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Ran An
- Department of Biochemistry and Biology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Yunfeng Li
- Department of Biochemistry and Biology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei Province, China
- The Seventh People's Hospital of Hebei Province, China
| |
Collapse
|
24
|
Ju JJ, Hang LH. Neuroinflammation and iron metabolism after intracerebral hemorrhage: a glial cell perspective. Front Neurol 2025; 15:1510039. [PMID: 39882361 PMCID: PMC11774705 DOI: 10.3389/fneur.2024.1510039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is the most common subtype of hemorrhagic stroke causing significant morbidity and mortality. Previously clinical treatments for ICH have largely been based on a single pathophysiological perspective, and there remains a lack of curative interventions. Following the rupture of cerebral blood vessels, blood metabolites activate resident immune cells such as microglia and astrocytes, and infiltrate peripheral immune cells, leading to the release of a series of inflammatory mediators. Degradation of hemoglobin produces large amounts of iron ions, leading to an imbalance of iron homeostasis and the production of large quantities of harmful hydroxyl radicals. Neuroinflammation and dysregulation of brain iron metabolism are both important pathophysiological changes in ICH, and both can exacerbate secondary brain injury. There is an inseparable relationship between brain iron metabolism disorder and activated glial cells after ICH. Glial cells participate in brain iron metabolism through various mechanisms; meanwhile, iron accumulation exacerbates neuroinflammation by activating inflammatory signaling pathways modulating the functions of inflammatory cells, and so on. This review aims to explore neuroinflammation from the perspective of iron metabolism, linking the complex pathophysiological changes, delving into the exploration of treatment approaches for ICH, and offering insights that could enhance clinical management strategies.
Collapse
Affiliation(s)
- Jia-Jun Ju
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
| | - Li-Hua Hang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
- Kunshan Cancer Pain Prevention and Treatment Key Laboratory, Kunshan, China
| |
Collapse
|
25
|
Liu M, Li B, Yin Z, Yin L, Luo Y, Zeng Q, Zhang D, Wu A, Chen L. Targeting mitochondrial dynamics: A promising approach for intracerebral hemorrhage therapy. Life Sci 2025; 361:123317. [PMID: 39674268 DOI: 10.1016/j.lfs.2024.123317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a major global health issue with high mortality and disability rates. Following ICH, the hematoma exerts direct pressure on brain tissue, and blood entering the brain directly damages neurons and the blood-brain barrier. Subsequently, oxidative stress, inflammatory responses, apoptosis, brain edema, excitotoxicity, iron toxicity, and metabolic dysfunction around the hematoma further exacerbate brain tissue damage, leading to secondary brain injury (SBI). Mitochondria, essential for energy production and the regulation of oxidative stress, are damaged after ICH, resulting in impaired ATP production, excessive reactive oxygen species (ROS) generation, and disrupted calcium homeostasis, all of which contribute to SBI. Therefore, a central factor in SBI is mitochondrial dysfunction. Mitochondrial dynamics regulate the shape, size, distribution, and quantity of mitochondria through fusion and fission, both of which are crucial for maintaining their function. Fusion repairs damaged mitochondria and preserves their health, while fission helps mitochondria adapt to cellular stress and removes damaged mitochondria through mitophagy. When this balance is disrupted following ICH, mitochondrial dysfunction worsens, oxidative stress and metabolic failure are exacerbated, ultimately contributing to SBI. Targeting mitochondrial dynamics offers a promising therapeutic approach to restoring mitochondrial function, reducing cellular damage, and improving recovery. This review explores the latest research on modulating mitochondrial dynamics and highlights its potential to enhance outcomes in ICH patients.
Collapse
Affiliation(s)
- Mengnan Liu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Binru Li
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China.
| | - Zhixue Yin
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Lu Yin
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Ye Luo
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Qi Zeng
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Dechou Zhang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Department of Cardiology, The Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Luzhou 646000, Sichuan, China; Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Li Chen
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
26
|
Wang R, Gan C, Mao R, Chen Y, Yan R, Li G, Xiong T, Guo J. Rat models of postintracerebral hemorrhage pneumonia induced by nasal inoculation with Klebsiella pneumoniae or intratracheal inoculation with LPS. Front Immunol 2025; 15:1477902. [PMID: 39845950 PMCID: PMC11750689 DOI: 10.3389/fimmu.2024.1477902] [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: 08/08/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background A stable and reproducible experimental bacterial pneumonia model postintracerebral hemorrhage (ICH) is necessary to help investigating the pathogenesis and novel treatments of Stroke-associated pneumonia (SAP). Aim To establish a Gram-negative bacterial pneumonia-complicating ICH rat model and an acute lung injury (ALI)-complicating ICH rat model. Methods We established two standardized models of post-ICH pneumonia by nasal inoculation with Klebsiella pneumoniae (Kp) or intratracheal inoculation with lipopolysaccharide (LPS). Survival and neurological scores were monitored. Magnetic resonance imaging was performed to evaluate hematoma volume. Abdominal aortic blood was collected for leukocyte counting, serum was isolated to determine concentrations of S100β and proinflammatory cytokines using ELISAs. Histopathological changes of brain, lung and gut were assessed using hematoxylin-eosin staining. Lung was isolated for immunofluorescence staining for myeloperoxidase (MPO). Bronchoalveolar lavage fluid was collected for leukocyte counting, and supernatant was prepared to measure MPO activity. Ileum was isolated for immunofluorescence staining for tight junction proteins ZO-1 and γδ TCRs/IL-17A and for Alcian blue-nuclear fast red staining of acidic mucins. Feces were collected, 16S rRNA sequencing, untargeted metabolomics and Spearman's correlation analyses were performed to explore changes of gut microbiota, metabolites and their interactions. Results In Kp-induced bacterial pneumonia-complicating ICH rats, we demonstrated that Kp challenge caused more severe neurological deficits, brain damage, neuroinflammation, and aggravated pneumonia and lung injury. Disruptions of the intestinal structure and gut barrier and the reductions of the protective intestinal IL-17A-producing γδT cells were also observed. Kp challenge exacerbated the gut microbiota dysbiosis and fecal metabolic profile disorders, which were characterized by abnormal sphingolipid metabolism especially elevated ceramide levels; increased levels of neurotoxic quinolinic acid and an upregulation of tryptophan (Trp)-serotonin-melatonin pathway. Spearman's correlation analyses further revealed that the reduction or depletion of some beneficial bacteria, such as Allobaculum and Faecalitalea, and the blooming of some opportunistic pathogens, such as Turicibacter, Dietzia, Corynebacterium and Clostridium_sensu_stricto_1 in Kp-induced SAP rats were associated with the disordered sphingolipid and Trp metabolism. Using an LPS-induced ALI complicating ICH model, we also characterized SAP-induced brain, lung and gut histopathology injuries; peripheral immune disorders and intense pulmonary inflammatory responses. Conclusions These two models may be highly useful for investigating the pathogenesis and screening and optimizing potential treatments for SAP. Moreover, the differential genera and sphingolipid or Trp metabolites identified above seem to be promising therapeutic targets.
Collapse
Affiliation(s)
- Ruihua Wang
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changlian Gan
- School of Traditional Dai Medicine, West Yunnan University of Applied Science, Xishuangbanna, China
| | - Rui Mao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Chen
- Department of Bioinformatics, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianqin Xiong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Neurology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
27
|
Zhao Q, Maimaitiaili S, Bi Y, Li M, Li X, Li Q, Shen X, Wu M, Fu L, Zhu Z, Zhang X, Chen J, Hu A, Zhang Z, Zhang W, Zhang B. Brain Iron Deposition Alterations in Type 2 Diabetes Mellitus Patients With Mild Cognitive Impairment Based on Quantitative Susceptibility Mapping. J Diabetes 2025; 17:e70052. [PMID: 39843980 PMCID: PMC11753919 DOI: 10.1111/1753-0407.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Iron is one of the most important elements in brain that may has a direct impact on the stability of central nervous system. The current study devoted to explore the alterations of iron distribution across the whole brain in type 2 diabetes mellitus (T2DM) patients with mild cognitive impairment (MCI). METHODS The quantitative susceptibility mapping (QSM) technique was used to quantify the intracranial iron content of 74 T2DM patients with MCI and 86 T2DM patients with normal cognition (NC). The group comparison was performed by a voxel-based analysis. Then we evaluated the relationships between cognitive indicators and magnetic susceptibility value (MSV) measured by QSM of the significant brain areas, which were set as the regions of interest (ROIs). In addition, we analyzed the moderation effects of grey matter volume (GMV) of the related brain areas and several metabolic and cerebrovascular factors on the associations between MSV of ROIs and cognitive characteristics. RESULTS T2DM patients with MCI exhibited a lower MSV in the right middle temporal gyrus (MTG) compared to NC group. And in the MCI group, there were significantly negative correlations between MSV of the right MTG and several memory indexes. Furthermore, the moderation effects of GMV of the whole brain and the bilateral MTG on the relationship between MSV of the right MTG and scores of list recognition were significant. CONCLUSIONS T2DM patients with MCI had a temporary decreased iron content in the right MTG, which may partially compensate for cognitive impairment. TRIAL REGISTRATION The study was registered at Clinicaltrials.gov (NCT02738671).
Collapse
Affiliation(s)
- Qiuyue Zhao
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Subinuer Maimaitiaili
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Yan Bi
- Department of Endocrinology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Ming Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Xin Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Qian Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Xinyi Shen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Min Wu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Linqing Fu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Zhengyang Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Anning Hu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Zhou Zhang
- Department of Endocrinology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Wen Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| |
Collapse
|
28
|
Zheng B, Zhou X, Pang L, Che Y, Qi X. Dihydroquercetin Ameliorates Neuronal Ferroptosis in Rats After Subarachnoid Hemorrhage via the PI3K/AKT/Nrf2/HO-1 Pathway. J Biochem Mol Toxicol 2025; 39:e70099. [PMID: 39756058 DOI: 10.1002/jbt.70099] [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: 05/17/2024] [Revised: 10/17/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025]
Abstract
Subarachnoid hemorrhage (SAH) is a specific type of stroke. Dihydroquercetin (DHQ), a flavonoid, is known for its various pharmacological properties. This study aimed to explore the roles and mechanisms of DHQ in influencing the progression of SAH. A rat SAH model was established using the endovascular perforation technique. Following SAH induction, DHQ was administered orally 1 h later. Assessments included SAH scores, neurological function, brain swelling, blood-brain barrier (BBB) integrity, neuronal damage, apoptosis levels, inflammation, and indicators of ferroptosis using various treatments. The HT22 cells were exposed to hemin to simulate SAH-like conditions under in vitro settings. Cell counting kit-8 assays, flow cytometry, enzyme?linked immunosorbent assay, BODIPY 581/591 C11 staining, western blot analysis, and biochemical kits were employed to evaluate the potential effects of DHQ. Moreover, the mechanisms responsible for the protective effect of DHQ were examined by western blot analysis. The in vivo findings revealed that DHQ mitigated neurological impairments, brain swelling, BBB disruption, and neuronal injury at 24 h post-SAH. DHQ also reduced neuronal degeneration, inflammation, and ferroptosis following SAH. The in vitro findings revealed that DHQ enhanced cell survival and reduced ferroptosis at 24 h following hemin exposure. Mechanistically, DHQ activated phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling in SAH rats and hemin-treated HT22 cells to exert neuroprotective effects. In conclusion, this study reveals that DHQ can effectively decrease BBB permeability, brain edema, neurological dysfunctions, and ferroptosis post-SAH by activating the PI3K/AKT/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Bao Zheng
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Xiwei Zhou
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Lujun Pang
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Yanjun Che
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Xin Qi
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| |
Collapse
|
29
|
Hao L, Zhang A, Lv D, Cong L, Sun Z, Liu L. EGCG activates Keap1/P62/Nrf2 pathway, inhibits iron deposition and apoptosis in rats with cerebral hemorrhage. Sci Rep 2024; 14:31474. [PMID: 39732956 PMCID: PMC11682079 DOI: 10.1038/s41598-024-82938-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: 06/28/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a common cerebrovascular disease characterized by a high incidence, disability rate, and mortality. Epigallocatechin gallate (EGCG), a key catechin compound found in green tea, has received increasing attention for its potential neuroprotective and therapeutic effects in neurological disorders. Studies have indicated that EGCG may influence various signaling pathways and molecular targets, including the inhibition of oxidative stress, reduction of inflammatory responses, suppression of cell apoptosis, regulation of cell survival, and enhancement of autophagy. Although the exact mechanism of action of EGCG is not fully understood, it has become a focal point of research in various disciplines due to its promising potential. This study aims to investigate the effects of EGCG on oxidative stress, iron deposition, and cell apoptosis in rats with ICH, as well as to uncover the underlying mechanisms. An ICH rat model was created to simulate cerebral hemorrhage, while an in vitro model utilizing primary cortical neurons was developed. The neurons were pre-treated with EGCG before being exposed to Erastin and RSL3 to induce iron death. The levels of oxidative stress, iron deposition, and cell apoptosis were evaluated in both models. In the ICH model, EGCG was discovered to enhance the activation of the Keap1/P62/Nrf2 signaling pathway in both in vivo and in vitro studies. Furthermore, EGCG significantly elevated the levels of GPX4 and XCT proteins, as well as the nuclear expression of Nrf2. It was noted that the Nrf2 inhibitor ML385 partially decreased the expression of these proteins. Through the activation of the Keap1/P62/Nrf2 pathway, EGCG inhibits inflammation, oxidative stress and iron deposition in rats with cerebral hemorrhage. EGCG inhibits oxidative stress, iron deposition and apoptosis in rats with ICH by activating Keap1/P62/Nrf2 pathway.
Collapse
Affiliation(s)
- Liang Hao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Department of Neurosurgery, The Third Hospital of Shijiazhuang, Shijiazhuang, 050000, Hebei, China
| | - Aobo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Dongsheng Lv
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - LuLu Cong
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zhimin Sun
- Department of Neurosurgery, The Third Hospital of Shijiazhuang, Shijiazhuang, 050000, Hebei, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
30
|
Feng M, An Y, Qin Q, Fong IH, Zhang K, Wang F, Song D, Li M, Yu M, Yeh CT, Chang J, Guo F. Sphk1/S1P pathway promotes blood-brain barrier breakdown after intracerebral hemorrhage through inducing Nlrp3-mediated endothelial cell pyroptosis. Cell Death Dis 2024; 15:926. [PMID: 39715736 DOI: 10.1038/s41419-024-07310-4] [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: 05/28/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024]
Abstract
Intracerebral hemorrhage (ICH) is a severe stroke subtype with high mortality and limited therapeutic options. The blood-brain barrier (BBB) breakdown post-ICH exacerbates secondary brain injury, highlighting the need for targeted therapies to preserve the BBB integrity. We aim to investigate the role of the Sphk1/S1P pathway in BBB breakdown following ICH and to evaluate the therapeutic potential of Sphk1 inhibition in mitigating this breakdown. Using a combination of human patient samples, mouse models of ICH, and in vitro cellular assays, we assessed the expression levels of Sphk1/S1P after ICH and changes of the BBB after ICH. The Sphk1 inhibitor PF543 and siRNAs were utilized to explore the pathway's impact on BBB integrity and the underlying mechanisms. The results indicate significant upregulation of Sphk1/S1P in the peri-hematomal brain tissue after ICH, which correlates with increased BBB leakage. Pharmacological inhibition of Sphk1 with PF543 attenuates BBB leakage, reduces hematoma volume, and improves neurological outcomes in mice. At the molecular and ultrastructural level, Sphk1 inhibition protects the BBB integrity by preserving tight junction proteins and suppressing endothelial transcytosis. Furthermore, mechanistic studies reveal that Sphk1 promotes Nlrp3-mediated pyroptosis of brain endothelial cells through the ERK1/2 signaling pathway. Taken together, the Sphk1/S1P pathway plays a critical role in ICH-induced BBB breakdown, and its inhibition represents a promising therapeutic strategy for ICH management.
Collapse
Affiliation(s)
- Mengzhao Feng
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
- Key Laboratory of Biomedical Imaging Science and System of Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, 518055, China
| | - Yuan An
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
- Key Laboratory of Biomedical Imaging Science and System of Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, 518055, China
| | - Qi Qin
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
- Key Laboratory of Biomedical Imaging Science and System of Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, 518055, China
| | - Iat-Hang Fong
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 23561, Taiwan
| | - Kaiyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
- Key Laboratory of Biomedical Imaging Science and System of Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, 518055, China
| | - Fang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
| | - Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
| | - Mengyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
| | - Min Yu
- Key Laboratory of Biomedical Imaging Science and System of Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, 518055, China.
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 23561, Taiwan.
- Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung, 95092, Taiwan.
| | - Junlei Chang
- Key Laboratory of Biomedical Imaging Science and System of Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, 518055, China.
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China.
| |
Collapse
|
31
|
Liu X, Wang W, Zhu F, Xu H, Ge G, Liang X, Yang H, Xu Y, Xu W, Wei M, Zhou Q, Geng D. Osteoblastic ferroptosis inhibition by small-molecule promoting GPX4 activation for peri-prosthetic osteolysis therapy. J Nanobiotechnology 2024; 22:758. [PMID: 39696565 DOI: 10.1186/s12951-024-03049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Peri-prosthesis osteolysis (PPO) represents the most severe complication of total joint arthroplasty (TJA) surgery and imposes the primary cause of prosthesis failure and subsequent revision surgery. Antiresorptive therapies are usually prescribed to treat PPO, especially for elderly people. Nevertheless, the efficacy of anti-osteoporotic medications remains constrained. Recent therapeutic strategies to promote periprosthetic osseointegration by restoring osteoblast function are considered more effective approaches. However, the precise mechanism underlying the inhibition of osteogenesis triggered by wear particles remains enigmatic. Herein, we demonstrate that wear particles inhibit osteoblast function by inducing ferroptosis to sabotage extracellular mineralization and arouse periprosthetic osteolysis. The suppression of ferroptosis could significantly rescue osteogenesis thus alleviating PPO. Furthermore, Glutathione Peroxidase 4 (GPX4) has been identified as a key target in regulating osteoblastic ferroptosis. By utilizing virtual screening techniques, we have successfully conducted a comprehensive screening of a natural compound known as Urolithin A (UA), which exhibits remarkable inhibition of osteoblastic ferroptosis while simultaneously promoting the process of osteogenesis through its precise targeting mechanism on GPX4. Meanwhile, UA improves the osteolytic conditions significantly in vivo even when the adjunction of titanium (Ti) nanoparticles. This strategy has great potential in treating peri-prosthesis osteolysis and potentially broadens the scope of clinical therapy.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wei Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Feng Zhu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Haibo Xu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Gaoran Ge
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaolong Liang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yaozeng Xu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wei Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Minggang Wei
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Qi Zhou
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200070, China.
| | - Dechun Geng
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
32
|
Mao J, Zhao Q, Guo M, Zhang S, Zhou J. Connecting the dots: Involvement of methyltransferase-like 3, N6-methyladenosine modification, and ferroptosis in the pathogenesis of intracerebral hemorrhage pathogenesis. Exp Neurol 2024; 382:114948. [PMID: 39260591 DOI: 10.1016/j.expneurol.2024.114948] [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: 05/29/2024] [Revised: 08/15/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024]
Abstract
Intracerebral hemorrhage is a profoundly detrimental acute cerebrovascular condition with a low overall survival rate and a high post-onset disability rate. Secondary brain injury that ensues post-ICH is the primary contributor to fatality and disability. Hence, the mitigation of brain injury during intracerebral hemorrhage progression has emerged as a crucial aspect of clinical management. N6-methyladenosine is the most pervasive, abundant, and conserved internal co-transcriptional modification of eukaryotic ribonucleic acid and is predominantly expressed in the nervous system. Methyltransferase-like 3 is a key regulatory protein that is strongly associated with the development of the nervous system and numerous neurological diseases. Ferroptosis, a form of iron-associated cell death, is a typical manifestation of neuronal apoptosis in neurological diseases and plays an important role in secondary brain damage following intracerebral hemorrhage. Therefore, this review aimed to elucidate the connection between m6A modification (particularly methyltransferase-like 3) and ferroptosis in the context of intracerebral hemorrhage to provide new insights for future intracerebral hemorrhage management approaches.
Collapse
Affiliation(s)
- Junxiang Mao
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Quantang Zhao
- Department of Neurosurgery, The Chinese People's Liberation Army Joint Logistics Support Force, No. 940 Hospital, Lanzhou City, Gansu Province, China
| | - Man Guo
- Department of Neurosurgery, The Chinese People's Liberation Army Joint Logistics Support Force, No. 940 Hospital, Lanzhou City, Gansu Province, China
| | - Shenghao Zhang
- Department of Neurosurgery, The Chinese People's Liberation Army Joint Logistics Support Force, No. 940 Hospital, Lanzhou City, Gansu Province, China
| | - Jie Zhou
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China.
| |
Collapse
|
33
|
Sun KY, Bai XY, Zhang L, Zhang X, Hu QQ, Song YX, Qiang RR, Zhang N, Zou JL, Yang YL, Xiang Y. A new strategy for the treatment of intracerebral hemorrhage: Ferroptosis. Exp Neurol 2024; 382:114961. [PMID: 39288829 DOI: 10.1016/j.expneurol.2024.114961] [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: 06/07/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Intracerebral hemorrhage, is a cerebrovascular disease with high morbidity, mortality, and disability. Due to the lack of effective clinical treatments, the development of new drugs to treat intracerebral hemorrhage is necessary. In recent years, ferroptosis has been found to play an important role in the pathophysiological process of intracerebral hemorrhage, which can be treated by inhibiting ferroptosis and thus intracerebral hemorrhage. This article aims to explain the mechanism of ferroptosis and its relationship to intracerebral hemorrhage. In the meantime, it briefly discusses the molecules identified to alleviate intracerebral hemorrhage by inhibiting ferroptosis, along with other clinical agents that are expected to treat intracerebral hemorrhage through this mechanism. In addition, a brief overview of the morphological alterations of different forms of cell death and their role in ICH is provided. Finally, the challenges that may arise in translating ferroptosis inhibitors from basic research to clinical use are presented. This article serves as a reference and provides insights to aid in the treatment of intracerebral hemorrhage in the clinic.
Collapse
Affiliation(s)
- Ke Yao Sun
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Qian Qian Hu
- School of Medicine, Yan'an University, Yan'an, China
| | - Yu Xuan Song
- School of Medicine, Yan'an University, Yan'an, China
| | | | - Ning Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Jia Lun Zou
- School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- School of Medicine, Yan'an University, Yan'an, China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, China; College of Physical Education, Yan'an University, Yan'an, China.
| |
Collapse
|
34
|
Fang H, Li M, Yang J, Ma S, Zhang L, Yang H, Tang Q, Cao J, Yang W. Repressing iron overload ameliorates central post-stroke pain via the Hdac2-Kv1.2 axis in a rat model of hemorrhagic stroke. Neural Regen Res 2024; 19:2708-2722. [PMID: 38595289 PMCID: PMC11168507 DOI: 10.4103/nrr.nrr-d-23-01498] [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: 09/06/2023] [Revised: 11/21/2023] [Accepted: 02/04/2024] [Indexed: 04/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202412000-00027/figure1/v/2024-04-08T165401Z/r/image-tiff Thalamic hemorrhage can lead to the development of central post-stroke pain. Changes in histone acetylation levels, which are regulated by histone deacetylases, affect the excitability of neurons surrounding the hemorrhagic area. However, the regulatory mechanism of histone deacetylases in central post-stroke pain remains unclear. Here, we show that iron overload leads to an increase in histone deacetylase 2 expression in damaged ventral posterolateral nucleus neurons. Inhibiting this increase restored histone H3 acetylation in the Kcna2 promoter region of the voltage-dependent potassium (Kv) channel subunit gene in a rat model of central post-stroke pain, thereby increasing Kcna2 expression and relieving central pain. However, in the absence of nerve injury, increasing histone deacetylase 2 expression decreased Kcna2 expression, decreased Kv current, increased the excitability of neurons in the ventral posterolateral nucleus area, and led to neuropathic pain symptoms. Moreover, treatment with the iron chelator deferiprone effectively reduced iron overload in the ventral posterolateral nucleus after intracerebral hemorrhage, reversed histone deacetylase 2 upregulation and Kv1.2 downregulation, and alleviated mechanical hypersensitivity in central post-stroke pain rats. These results suggest that histone deacetylase 2 upregulation and Kv1.2 downregulation, mediated by iron overload, are important factors in central post-stroke pain pathogenesis and could serve as new targets for central post-stroke pain treatment.
Collapse
Affiliation(s)
- He Fang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingchen Yang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shunping Ma
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Li Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hongqi Yang
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, Henan Province, China
| | - Qiongyan Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, Henan Province, China
| | - Weimin Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
35
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
36
|
Tao J, Li J, Fan X, Jiang C, Wang Y, Qin M, Nikfard Z, Nikfard F, Wang Y, Zhao T, Xing N, Zille M, Wang J, Zhang J, Chen X, Wang J. Unraveling the protein post-translational modification landscape: Neuroinflammation and neuronal death after stroke. Ageing Res Rev 2024; 101:102489. [PMID: 39277050 DOI: 10.1016/j.arr.2024.102489] [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: 08/07/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
The impact of stroke on global health is profound, with both high mortality and morbidity rates. This condition can result from cerebral ischemia, intracerebral hemorrhage (ICH), and subarachnoid hemorrhage (SAH). The pathophysiology of stroke involves secondary damage and irreversible loss of neuronal function. Post-translational modifications (PTMs) have been recognized as crucial regulatory mechanisms in ischemic and hemorrhagic stroke-induced brain injury. These PTMs include phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and succinylation. This comprehensive review delves into recent research on the PTMs landscape associated with neuroinflammation and neuronal death specific to cerebral ischemia, ICH, and SAH. This review aims to explain the role of PTMs in regulating pathologic mechanisms and present critical techniques and proteomic strategies for identifying PTMs. This knowledge helps us comprehend the underlying mechanisms of stroke injury and repair processes, leading to the development of innovative treatment strategies. Importantly, this review underscores the significance of exploring PTMs to understand the pathophysiology of stroke.
Collapse
Affiliation(s)
- Jin Tao
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Jiaxin Li
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China
| | - Yebin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Mengzhe Qin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Zahra Nikfard
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Fatemeh Nikfard
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Yunchao Wang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, P. R. China
| | - Ting Zhao
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P. R. China
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| |
Collapse
|
37
|
Zheng Y, Chen X, Wang Y, Chen Z, Wu D. Phenolic-enabled nanotechnology: a new strategy for central nervous system disease therapy. J Zhejiang Univ Sci B 2024; 25:890-913. [PMID: 39420524 PMCID: PMC11494163 DOI: 10.1631/jzus.b2300839] [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: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 10/19/2024]
Abstract
Polyphenolic compounds have received tremendous attention in biomedicine because of their good biocompatibility and unique physicochemical properties. In recent years, phenolic-enabled nanotechnology (PEN) has become a hotspot of research in the medical field, and many promising studies have been reported, especially in the application of central nervous system (CNS) diseases. Polyphenolic compounds have superior anti-inflammatory and antioxidant properties, and can easily cross the blood‒brain barrier, as well as protect the nervous system from metabolic damage and promote learning and cognitive functions. However, although great advances have been made in this field, a comprehensive review regarding PEN-based nanomaterials for CNS therapy is lacking. A systematic summary of the basic mechanisms and synthetic strategies of PEN-based nanomaterials is beneficial for meeting the demand for the further development of novel treatments for CNS diseases. This review systematically introduces the fundamental physicochemical properties of PEN-based nanomaterials and their applications in the treatment of CNS diseases. We first describe the different ways in which polyphenols interact with other substances to form high-quality products with controlled sizes, shapes, compositions, and surface chemistry and functions. The application of PEN-based nanomaterials in the treatment of CNS diseases is then described, which provides a reference for subsequent research on the treatment of CNS diseases.
Collapse
Affiliation(s)
- Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. ,
- Zhejiang Rehabilitation Medical Center, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310009, China. ,
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. ,
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
38
|
Boren SB, Savitz SI, Gonzales N, Hasan K, Becerril-Gaitan A, Maroufy V, Li Y, Grotta J, Steven EA, Chen CJ, Sitton CW, Aronowski J, Haque ME. Longitudinal Morphometric Changes in the Corticospinal Tract Shape After Hemorrhagic Stroke. Transl Stroke Res 2024; 15:893-901. [PMID: 37308620 DOI: 10.1007/s12975-023-01168-y] [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: 05/17/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Deep intracerebral hemorrhage (ICH) exerts a direct force on corticospinal tracts (CST) causing shape deformation. Using serial MRI, Generalized Procrustes Analysis (GPA), and Principal Components Analysis (PCA), we temporally evaluated the change in CST shape. Thirty-five deep ICH patients with ipsilesional-CST deformation were serially imaged on a 3T-MRI with a median imaging time of day-2 and 84 of onset. Anatomical and diffusion tensor images (DTI) were acquired. Using DTI color-coded maps, 15 landmarks were drawn on each CST and the centroids were computed in 3 dimensions. The contralesional-CST landmarks were used as a reference. The GPA outlined the shape coordinates and we superimposed the ipsilesional-CST shape at the two-time points. A multivariate PCA was applied to identify eigenvectors associated with the highest percentile of change. The first three principal components representing CST deformation along the left-right (PC1), anterior-posterior (PC2), and superior-inferior (PC3) respectively were responsible for 57.9% of shape variance. The PC1 (36.1%, p < 0.0001) and PC3 (9.58%, p < 0.01) showed a significant deformation between the two-time points. Compared to the contralesional-CST, the ipsilesional PC scores were significantly (p < 0.0001) different only at the first-timepoint. A significant positive association between the ipsilesional-CST deformation and hematoma volume was observed. We present a novel method to quantify CST deformation caused by ICH. Deformation most often occurs in left-right axis (PC1) and superior-inferior (PC3) directions. As compared to the reference, the significant temporal difference at the first time point suggests CST restoration over time.
Collapse
Affiliation(s)
- Seth B Boren
- Institute for Stroke and Cerebrovascular Diseases and Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Sean I Savitz
- Institute for Stroke and Cerebrovascular Diseases and Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Nicole Gonzales
- Institute for Stroke and Cerebrovascular Diseases and Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77030, USA
- Department of Neurology, Neurohospitalist and Stroke Section, University of Colorado School of Medicine, Aurora, USA
| | - Khader Hasan
- Department of Interventional Diagnostic Radiology, McGovern Medical School, The University of Texas Health Science Center, Houston, USA
| | - Andrea Becerril-Gaitan
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center, Houston, USA
| | - Vahed Maroufy
- Department of Biostatistics and Data Science, School of Public Health, McGovern Medical School, The University of Texas Health Science Center, Houston, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, School of Public Health, McGovern Medical School, The University of Texas Health Science Center, Houston, USA
| | - James Grotta
- Stroke Research and Mobile Stroke Unit, Department of Neurology, Memorial Hermann Hospital, Houston, USA
| | - Emily A Steven
- Institute for Stroke and Cerebrovascular Diseases and Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Ching-Jen Chen
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center, Houston, USA
| | - Clark W Sitton
- Department of Interventional Diagnostic Radiology, McGovern Medical School, The University of Texas Health Science Center, Houston, USA
| | - Jaroslaw Aronowski
- Institute for Stroke and Cerebrovascular Diseases and Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Muhammad E Haque
- Institute for Stroke and Cerebrovascular Diseases and Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77030, USA.
| |
Collapse
|
39
|
Han J, Zhang J, Yao X, Meng M, Wan Y, Cheng Y. Mechanism of HDAC1 Regulating Iron Overload-Induced Neuronal Oxidative Damage After Cerebral Hemorrhage. Mol Neurobiol 2024; 61:7549-7566. [PMID: 38403721 DOI: 10.1007/s12035-024-04000-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
Iron overload is associated with brain edema in the context of intracerebral hemorrhage (ICH). Here, we investigated the role of histone deacetylase 1 (HDAC1) in mediating oxidative damage induced by iron overload after ICH. Utilizing ICH mouse models and FeCl2-induced HT-22 cell models, we assessed HDAC1 expression and its impact on iron overload and oxidative damage. We examined the levels of Kruppel like factor 4 (KLF4), RAN binding protein 9 (RANBP9), as well as the acetylation levels of HDAC1 and histones H3 and H4 in the KLF4 promoter, and the KLF4 level in the RANBP9 promoter. Additionally, we investigated the binding relationships between KLF4 and the RANBP9 promoter, HDAC1 and miR-129-5p. Our results demonstrated elevated HDAC1 expression in ICH mice and FeCl2-induced HT-22 cells. HDAC1 silencing improved neurological function in mice, reduced brain edema, and alleviated iron overload and oxidative damage in vitro. HDAC1 downregulated KLF4 expression by reducing acetylation levels in the KLF4 promoter, leading to decreased KLF4 enrichment in the RANBP9 promoter and increased RANBP9 expression. Furthermore, upstream miR-129-5p inhibited HDAC1, and the downregulation of miR-129-5p mitigated the protective effect of HDAC1 silencing. Collectively, our findings highlight the significant role of HDAC1 in exacerbating iron overload-induced oxidative damage following ICH and its regulation by miR-129-5p.
Collapse
Affiliation(s)
- Jing Han
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jinnan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xiaojuan Yao
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Meng Meng
- Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, 300000, China
| | - Yahui Wan
- Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, 300000, China
| | - Yan Cheng
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
40
|
Sun Y, Sun W, Liu J, Zhang B, Zheng L, Zou W. The dual role of microglia in intracerebral hemorrhage. Behav Brain Res 2024; 473:115198. [PMID: 39128628 DOI: 10.1016/j.bbr.2024.115198] [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: 03/15/2024] [Revised: 07/05/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
Intracerebral hemorrhage has the characteristics of high morbidity, disability and mortality, which has caused a heavy burden to families and society. Microglia are resident immune cells in the central nervous system, and their activation plays a dual role in tissue damage after intracerebral hemorrhage. The damage in cerebral hemorrhage is embodied in the following aspects: releasing inflammatory factors and inflammatory mediators, triggering programmed cell death, producing glutamate induced excitotoxicity, and destroying blood-brain barrier; The protective effect is reflected in the phagocytosis and clearance of harmful substances by microglia, and the secretion of anti-inflammatory and neurotrophic factors. This article summarizes the function of microglia and its dual regulatory mechanism in intracerebral hemorrhage. In the future, drugs, acupuncture and other clinical treatments can be used to intervene in the activation state of microglia, so as to reduce the harm of microglia.
Collapse
Affiliation(s)
- Yue Sun
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Wentao Sun
- Faculty of Chinese Medicine Sciense Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China
| | - Jiawei Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Baiwen Zhang
- Clinical Key Laboratory of Integrated Traditional Chinese and Western Medicine of Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Lei Zheng
- Clinical Key Laboratory of Integrated Traditional Chinese and Western Medicine of Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China.
| |
Collapse
|
41
|
Alsharabasy AM, Lagarias PI, Papavasileiou KD, Afantitis A, Farràs P, Glynn S, Pandit A. Examining Hemin and its Derivatives: Induction of Heme-Oxygenase-1 Activity and Oxidative Stress in Breast Cancer Cells through Collaborative Experimental Analysis and Molecular Dynamics Simulations. J Med Chem 2024; 67:15411-15427. [PMID: 39159487 PMCID: PMC11403666 DOI: 10.1021/acs.jmedchem.4c00989] [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: 08/21/2024]
Abstract
Hemin triggers intracellular reactive oxygen species (ROS) accumulation and enhances heme oxygenase-1 (HOX-1) activity, indicating its potential as an anticancer agent, though precise control of its intracellular levels is crucial. The study explores the impact of hemin and its derivatives, hemin-tyrosine, and hemin-styrene (H-Styr) conjugates on migration, HOX-1 expression, specific apoptosis markers, mitochondrial functions, and ROS generation in breast cancer cells. Molecular docking and dynamics simulations were used to understand the interactions among HOX-1, heme, and the compounds. Hemin outperforms its derivatives in inducing HOX-1 expression, exhibiting pro-oxidative effects and reducing cell migration. Molecular simulations show that heme binds favorably to HOX-1, followed by the other compounds, primarily through van der Waals and electrostatic forces. However, only van der Waals forces determine the H-Styr complexation. These interactions, influenced by metalloporphyrin characteristics, provide insights into HOX-1 regulation and ROS generation, potentially guiding the development of breast cancer therapies targeting oxidative stress.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | | | - Konstantinos D Papavasileiou
- Department of ChemoInformatics, Novamechanics Ltd., Nicosia 1070, Cyprus
- Department of Chemoinformatics, Novamechanics MIKE, Piraeus 18545, Greece
- Division of Data Driven Innovation, Entelos Institute, Larnaca 6059, Cyprus
| | - Antreas Afantitis
- Department of ChemoInformatics, Novamechanics Ltd., Nicosia 1070, Cyprus
- Department of Chemoinformatics, Novamechanics MIKE, Piraeus 18545, Greece
- Division of Data Driven Innovation, Entelos Institute, Larnaca 6059, Cyprus
| | - Pau Farràs
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- School of Biological and Chemical Sciences, Ryan Institute, University of Galway, Galway H91 TK33, Ireland
| | - Sharon Glynn
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway H91 YR71, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
42
|
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
|
43
|
Karatoprak DE, Engin R, Sahin S, İclek İ, Durak MA. Investigation of Neuroprotective Efficacy of Dexpanthenol in an Experimental Head Injury Model. J Korean Neurosurg Soc 2024; 67:521-530. [PMID: 38449284 PMCID: PMC11375073 DOI: 10.3340/jkns.2023.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/16/2023] [Indexed: 03/08/2024] Open
Abstract
OBJECTIVE Dexpanthenol (DXP), which has known neuroprotective effects, has been shown to be beneficial in various experimental models and ischaemic diseases. The aim of this study was to investigate the possible neuroprotective effects of DXP in a traumatic brain injury (TBI) model. METHODS Thirty-six Wistar-Albino female rats, approximately 6 months old, weighing 220-285 g were used. All rats were subjected to closed head trauma by dropping a weight of 350 g on the parietal region from a height of 50 cm at an angle of 180 degrees in the prepared head trauma model setup. The rats were divided into four groups as control (group 1), trauma (group 2), trauma + DXP (group 3), and DXP (group 4). In group 3, DXP was administered intraperitoneally at a dose of 500 mg/kg for six times at 30 minutes, 6, 12, 24, 36, and 48 hours. In group 4, DXP was administered intraperitoneally simultaneously with group 3 without causing head trauma. Blood samples were taken from all rats 72 hours later for biochemical examination. After blood samples were taken, rats were decapitated under general anaesthesia. Cerebral tissue samples were taken from decapitated rats for immunohistochemical and histopathological examination. RESULTS Cytokine markers were found to be increased in posttraumatic brain tissue. Malondialdehyde and glutathione reductase levels were lower in group 3 compared to group 2. In addition, superoxide dismutase, glutathione peroxidase and catalase levels were significantly higher in group 3 compared to group 2. In histological evaluation, congestion in the piamater layer, cell infiltration, vascular congestion, hemorrhage and neuronal degeneration were significantly decreased in group 3 compared to group 2. DXP seems to be beneficial in neurological recovery in terms of histological and oxidative changes after head trauma in rats. CONCLUSION DXP should be further evaluated for its possible therapeutic effect in TBI.
Collapse
Affiliation(s)
- Durmus E. Karatoprak
- Brain and Nerve Surgery Clinic, Ministry of Health Necip Fazıl City Hospital, Kahramanmaraş, Turkey
| | - Recai Engin
- Brain and Nerve Surgery Clinic, Ministry of Health Necip Fazıl City Hospital, Kahramanmaraş, Turkey
| | - Sarp Sahin
- Brain and Nerve Surgery Clinic, Ministry of Health Karadeniz Ereğli State Hospital, Zonguldak, Turkey
| | - İsmail İclek
- Department of Neurosurgery, Inonu University Faculty of Medicine, Malatya, Turkey
| | - Mehmet A. Durak
- Department of Neurosurgery, Inonu University Faculty of Medicine, Malatya, Turkey
| |
Collapse
|
44
|
Boinska J, Słomka A, Sury M, Wiszniewska M, Pisarek E, Żekanowska E. Insights into Iron Metabolism Parameters in Ischemic Stroke: A Single-Center Prospective Cohort Study. Int J Mol Sci 2024; 25:9352. [PMID: 39273300 PMCID: PMC11395666 DOI: 10.3390/ijms25179352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
The hemojuvelin-hepcidin regulatory axis may play a key role in the iron metabolism both systemically and locally. There is a pressing need to evaluate this tightly regulated network of iron parameters and their potential impact on the development of ischemic stroke (IS). We aimed to assess iron metabolism biomarkers in patients after IS, evaluating changes over time and considering their clinical features. We studied 45 patients diagnosed with IS. We assessed major iron metabolism parameters, such as hepcidin, soluble hemojuvelin (sHJV), soluble transferrin receptor (sTfR), and ferritin, using immunoenzymathic methods at two time points: on admission and on the 7th day post IS. We found increased ferritin levels on the 7th day post IS compared to admission, and this was observed in the entire study group (p = 0.03) and in the subgroup treated with thrombolysis (p = 0.02). The hepcidin levels, on the other hand, showed a significant decrease on the 7th day, though this difference was only evident in the entire study group (p = 0.04). We also discovered significantly elevated sHJV levels in patients with PACI stroke compared to other stroke locations, both on admission and on the 7th day post IS (p < 0.05). Significantly higher sHJV levels were observed in patients treated with thrombolysis compared to those receiving conventional treatment, regardless of the time point (p < 0.0001 and p = 0.0002, respectively). Our study revealed changes in the iron metabolism parameters during stroke. The patients with anterior cerebral infarction and those treated with thrombolysis presented significantly elevated sHJV levels.
Collapse
Affiliation(s)
- Joanna Boinska
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 Marii Curie-Skłodowskiej Street, 85-094 Bydgoszcz, Poland
| | - Artur Słomka
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 Marii Curie-Skłodowskiej Street, 85-094 Bydgoszcz, Poland
| | - Magdalena Sury
- Neurological Department with Stroke Unit, Specialist Hospital, 64-920 Piła, Poland
| | - Małgorzata Wiszniewska
- Neurological Department with Stroke Unit, Specialist Hospital, 64-920 Piła, Poland
- Faculty of Nursing, Stanisław Staszic State University of Applied Sciences, 64-920 Piła, Poland
| | - Ewa Pisarek
- Faculty of Nursing, Stanisław Staszic State University of Applied Sciences, 64-920 Piła, Poland
| | - Ewa Żekanowska
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 Marii Curie-Skłodowskiej Street, 85-094 Bydgoszcz, Poland
| |
Collapse
|
45
|
Wróblewski M, Wróblewska W, Sobiesiak M. The Role of Selected Elements in Oxidative Stress Protection: Key to Healthy Fertility and Reproduction. Int J Mol Sci 2024; 25:9409. [PMID: 39273356 PMCID: PMC11395468 DOI: 10.3390/ijms25179409] [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: 08/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Oxidative stress and its relationship to fertility and reproduction is a topic of interest in medicine, especially in the context of the effects of trace elements and micronutrients. Oxidative stress occurs when there is an excess of free radicals in the body, which can lead to cell and tissue damage. Free radicals are reactive oxygen species (ROS) that can be formed as a result of normal metabolic processes, as well as under the influence of external factors such as environmental pollution, UV radiation, and diet. Oxidative stress has a significant impact on fertility. In men, it can lead to DNA damage in sperm, which can result in reduced semen quality, reduced sperm motility and increased numbers of defective sperm, and free radical damage to sperm cell membranes causing a reduction in the number of available sperm. In women, oxidative stress can affect the quality of female reproductive cells, which can lead to problems with their maturation and with embryo implantation in the uterus and can also affect ovarian function and disrupt hormonal regulation of the menstrual cycle. A proper balance of trace elements and micronutrients is key to protecting against oxidative stress and maintaining reproductive health. Supplementation with appropriate elements such as zinc, selenium, copper, manganese, chromium, and iron can help reduce oxidative stress and improve fertility. This work discusses the effects of selected elements on oxidative stress parameters specifically in terms of fertility and reproduction.
Collapse
Affiliation(s)
- Marcin Wróblewski
- Department of Medical Biology and Biochemistry, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Weronika Wróblewska
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Marta Sobiesiak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| |
Collapse
|
46
|
Wang W, Liu X, Wang Y, Zhou D, Chen L. Application of biomaterials in the treatment of intracerebral hemorrhage. Biomater Sci 2024; 12:4065-4082. [PMID: 39007343 DOI: 10.1039/d4bm00630e] [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: 07/16/2024]
Abstract
Although the current surgical hematoma removal treatment saves patients' lives in critical moments of intracerebral hemorrhage (ICH), the lethality and disability rates of ICH are still very high. Due to the individual differences of patients, postoperative functional improvement is still to be confirmed, and the existing drug treatment has limited benefits for ICH. Recent advances in biomaterials may provide new ideas for the therapy of ICH. This review first briefly describes the pathogenic mechanisms of ICH, including primary and secondary injuries such as inflammation and intracerebral edema, and briefly describes the existing therapeutic approaches and their limitations. Secondly, existing nanomaterials and hydrogels for ICH, including exosomes, liposomes, and polymer nanomaterials, are also described. In addition, the potential challenges and application prospects of these biomaterials for clinical translation in ICH treatment are discussed.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510310, P. R. China.
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China.
| | - Xiaowen Liu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510310, P. R. China.
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China.
| | - Yupeng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China.
| | - Dongfang Zhou
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510310, P. R. China.
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China.
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510310, P. R. China.
| |
Collapse
|
47
|
Pustimbara A, Li C, Ogura SI. Hemin enhances the 5-aminolevulinic acid-photodynamic therapy effect through the changes of cellular iron homeostasis. Photodiagnosis Photodyn Ther 2024; 48:104253. [PMID: 38901716 DOI: 10.1016/j.pdpdt.2024.104253] [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: 05/17/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Photodynamic therapy (PDT) has been utilized as a promising alternative cancer treatment due to its minimum invasiveness over the years. Exogenous 5-aminolevulinic acid (ALA) triggers protoporphyrin IX (PpIX) accumulation, which happens in cancer cells. However, certain types of cancer exhibit reduced effectiveness in the PpIX accumulation mechanism. This study aimed to determine the effect of ALA-PDT combination with hemin on gastric carcinoma TMK-1 cells. METHODS This study utilized TMK-1 gastric cancer cell line to evaluate PpIX, ROS, and Fe2+ accumulation following the administration of ALA, hemin, and a combination of ALA and hemin PDT. We also evaluate the mRNA expressions related to iron homeostasis and treatment impacts on cell viability. RESULTS The co-addition of ALA and hemin PDT for 4 h of treatment resulted in a significant decrease in cell viability by up to 18 %. While ALA-PDT enhanced PpIX metabolism, the addition of hemin influenced both the production of reactive oxygen species (ROS) and cellular iron homeostasis by inducing Fe2+ accumulation and affecting mRNA levels of IRP, Tfr1, Ferritin, NFS1, and SDHB. CONCLUSION These findings suggest that the addition of ALA and hemin enhances phototoxicity in TMK-1 cells. The combination of ALA and hemin with PDT induces cell death, evidenced by increased cytotoxicity properties such as PpIX and ROS, along with significant changes in TMK-1 gastric cancer iron homeostasis. Therefore, the combination of ALA and hemin could be one of the alternatives in photodynamic therapy for cancer in the future.
Collapse
Affiliation(s)
- Anantya Pustimbara
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| | - Chenhan Li
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| | - Shun-Ichiro Ogura
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| |
Collapse
|
48
|
Cong J, Li JY, Zou W. Mechanism and treatment of intracerebral hemorrhage focus on mitochondrial permeability transition pore. Front Mol Neurosci 2024; 17:1423132. [PMID: 39156127 PMCID: PMC11328408 DOI: 10.3389/fnmol.2024.1423132] [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: 04/25/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is the second most common subtype of stroke, characterized by high mortality and a poor prognosis. Despite various treatment methods, there has been limited improvement in the prognosis of ICH over the past decades. Therefore, it is imperative to identify a feasible treatment strategy for ICH. Mitochondria are organelles present in most eukaryotic cells and serve as the primary sites for aerobic respiration and energy production. Under unfavorable cellular conditions, mitochondria can induce changes in permeability through the opening of the mitochondrial permeability transition pore (mPTP), ultimately leading to mitochondrial dysfunction and contributing to various diseases. Recent studies have demonstrated that mPTP plays a role in the pathological processes associated with several neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, Huntington's disease, ischemic stroke and ischemia-reperfusion injury, among others. However, there is limited research on mPTP involvement specifically in ICH. Therefore, this study comprehensively examines the pathological processes associated with mPTP in terms of oxidative stress, apoptosis, necrosis, autophagy, ferroptosis, and other related mechanisms to elucidate the potential mechanism underlying mPTP involvement in ICH. This research aims to provide novel insights for the treatment of secondary injury after ICH.
Collapse
Affiliation(s)
- Jing Cong
- The First School of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing-Yi Li
- The Second School of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Zou
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
49
|
Zeng H, Zeng D, Yin X, Zhang W, Wu M, Chen Z. Research progress on high-concentration oxygen therapy after cerebral hemorrhage. Front Neurol 2024; 15:1410525. [PMID: 39139771 PMCID: PMC11320605 DOI: 10.3389/fneur.2024.1410525] [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: 04/01/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Recently, the role of high-concentration oxygen therapy in cerebral hemorrhage has been extensively discussed. This review describes the research progress in high-concentration oxygen therapy after cerebral hemorrhage. High-concentration oxygen therapy can be classified into two treatment methods: hyperbaric and normobaric high-concentration oxygen therapy. Several studies have reported that high-concentration oxygen therapy uses the pathological mechanisms of secondary ischemia and hypoxia after cerebral hemorrhage as an entry point to improve cerebral oxygenation, metabolic rate, cerebral edema, intracranial pressure, and oxidative stress. We also elucidate the mechanisms by which molecules such as Hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor, and erythropoietin (EPO) may play a role in oxygen therapy. Although people are concerned about the toxicity of hyperoxia, combined with relevant literature, the evidence discussed in this article suggests that as long as the duration, concentration, pressure, and treatment interval of patients with cerebral hemorrhage are properly understood and oxygen is administered within the treatment window, it can be effective to avoid hyperoxic oxygen toxicity. Combined with the latest research, we believe that high-concentration oxygen therapy plays an important positive role in injuries and outcomes after cerebral hemorrhage, and we recommend expanding the use of normal-pressure high-concentration oxygen therapy for cerebral hemorrhage.
Collapse
Affiliation(s)
- He Zeng
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Dakai Zeng
- Department of Anorectal Surgery, Third Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Wumiao Zhang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Moxin Wu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
50
|
Sahebi K, Foroozand H, Amirsoleymani M, Eslamzadeh S, Negahdaripour M, Tajbakhsh A, Rahimi Jaberi A, Savardashtaki A. Advancing stroke recovery: unlocking the potential of cellular dynamics in stroke recovery. Cell Death Discov 2024; 10:321. [PMID: 38992073 PMCID: PMC11239950 DOI: 10.1038/s41420-024-02049-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 07/13/2024] Open
Abstract
Stroke stands as a predominant cause of mortality and morbidity worldwide, and there is a pressing need for effective therapies to improve outcomes and enhance the quality of life for stroke survivors. In this line, effective efferocytosis, the clearance of apoptotic cells, plays a crucial role in neuroprotection and immunoregulation. This process involves specialized phagocytes known as "professional phagocytes" and consists of four steps: "Find-Me," "Eat-Me," engulfment/digestion, and anti-inflammatory responses. Impaired efferocytosis can lead to secondary necrosis and inflammation, resulting in adverse outcomes following brain pathologies. Enhancing efferocytosis presents a potential avenue for improving post-stroke recovery. Several therapeutic targets have been identified, including osteopontin, cysteinyl leukotriene 2 receptor, the µ opioid receptor antagonist β-funaltrexamine, and PPARγ and RXR agonists. Ferroptosis, defined as iron-dependent cell death, is now emerging as a novel target to attenuate post-stroke tissue damage and neuronal loss. Additionally, several biomarkers, most importantly CD163, may serve as potential biomarkers and therapeutic targets for acute ischemic stroke, aiding in stroke diagnosis and prognosis. Non-pharmacological approaches involve physical rehabilitation, hypoxia, and hypothermia. Mitochondrial dysfunction is now recognized as a major contributor to the poor outcomes of brain stroke, and medications targeting mitochondria may exhibit beneficial effects. These strategies aim to polarize efferocytes toward an anti-inflammatory phenotype, limit the ingestion of distressed but viable neurons, and stimulate efferocytosis in the late phase of stroke to enhance post-stroke recovery. These findings highlight promising directions for future research and development of effective stroke recovery therapies.
Collapse
Affiliation(s)
- Keivan Sahebi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Foroozand
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Saghi Eslamzadeh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Abbas Rahimi Jaberi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|