1
|
Yang J, Wu J, Xie X, Xia P, Lu J, Liu J, Bai L, Li X, Yu Z, Li H. Perilipin-2 mediates ferroptosis in oligodendrocyte progenitor cells and myelin injury after ischemic stroke. Neural Regen Res 2025; 20:2015-2028. [PMID: 39254564 DOI: 10.4103/nrr.nrr-d-23-01540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/27/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00024/figure1/v/2024-09-09T124005Z/r/image-tiff Differentiation of oligodendrocyte progenitor cells into mature myelin-forming oligodendrocytes contributes to remyelination. Failure of remyelination due to oligodendrocyte progenitor cell death can result in severe nerve damage. Ferroptosis is an iron-dependent form of regulated cell death caused by membrane rupture induced by lipid peroxidation, and plays an important role in the pathological process of ischemic stroke. However, there are few studies on oligodendrocyte progenitor cell ferroptosis. We analyzed transcriptome sequencing data from GEO databases and identified a role of ferroptosis in oligodendrocyte progenitor cell death and myelin injury after cerebral ischemia. Bioinformatics analysis suggested that perilipin-2 (PLIN2) was involved in oligodendrocyte progenitor cell ferroptosis. PLIN2 is a lipid storage protein and a marker of hypoxia-sensitive lipid droplet accumulation. For further investigation, we established a mouse model of cerebral ischemia/reperfusion. We found significant myelin damage after cerebral ischemia, as well as oligodendrocyte progenitor cell death and increased lipid peroxidation levels around the infarct area. The ferroptosis inhibitor, ferrostatin-1, rescued oligodendrocyte progenitor cell death and subsequent myelin injury. We also found increased PLIN2 levels in the peri-infarct area that co-localized with oligodendrocyte progenitor cells. Plin2 knockdown rescued demyelination and improved neurological deficits. Our findings suggest that targeting PLIN2 to regulate oligodendrocyte progenitor cell ferroptosis may be a potential therapeutic strategy for rescuing myelin damage after cerebral ischemia.
Collapse
Affiliation(s)
- Jian Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Xueshun Xie
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Pengfei Xia
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiale Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Wang Y, Yuan T, Lyu T, Zhang L, Wang M, He Z, Wang Y, Li Z. Mechanism of inflammatory response and therapeutic effects of stem cells in ischemic stroke: current evidence and future perspectives. Neural Regen Res 2025; 20:67-81. [PMID: 38767477 PMCID: PMC11246135 DOI: 10.4103/1673-5374.393104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/13/2023] [Accepted: 11/21/2023] [Indexed: 05/22/2024] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide, with an increasing trend and tendency for onset at a younger age. China, in particular, bears a high burden of stroke cases. In recent years, the inflammatory response after stroke has become a research hotspot: understanding the role of inflammatory response in tissue damage and repair following ischemic stroke is an important direction for its treatment. This review summarizes several major cells involved in the inflammatory response following ischemic stroke, including microglia, neutrophils, monocytes, lymphocytes, and astrocytes. Additionally, we have also highlighted the recent progress in various treatments for ischemic stroke, particularly in the field of stem cell therapy. Overall, understanding the complex interactions between inflammation and ischemic stroke can provide valuable insights for developing treatment strategies and improving patient outcomes. Stem cell therapy may potentially become an important component of ischemic stroke treatment.
Collapse
Affiliation(s)
- Yubo Wang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tingli Yuan
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
| | - Tianjie Lyu
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Meng Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhiying He
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yongjun Wang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Zixiao Li
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| |
Collapse
|
3
|
Chen X, Wang L, Wang N, Li C, Hang H, Wu G, Ren S, Jun T, Wang L. An apolipoprotein E receptor mimetic peptide decreases blood-brain barrier permeability following intracerebral hemorrhage by inhibiting the CypA/MMP-9 signaling pathway via LRP1 activation. Int Immunopharmacol 2024; 143:113007. [PMID: 39486173 DOI: 10.1016/j.intimp.2024.113007] [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: 06/10/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 11/04/2024]
Abstract
Apolipoprotein (Apo) E mimetic peptides down-regulate the inflammatory response and alleviate damage to secondary neurons after intracerebral hemorrhage (ICH). We designed a novel apoE receptor mimetic composed of the low-density lipoprotein receptor-associated protein-1 (LRP1) receptor-binding domain of apoE with 6 lysines (6KApoEp). The 6KApoEp peptide is small enough to penetrate the blood-brain barrier (BBB) and modulate the inflammatory response during damage to the central nervous system. LRP1 inhibits the CypA/MMP-9 pathway and reduces BBB damage. Thus, we examined the effects of 6KApoEp-LRP1 interaction. LRP1 and 6KApoEp interacted and co-localized in the pericytes. We established a Sprague-Dawley (SD) male rat model of ICH to observe the role of 6KApoEp in secondary injury after ICH. The expression levels of cyclophilin A (CypA), nuclear factor kappa-B (NF-κB) p65, and matrix metalloproteinase 9 (MMP-9) were increased, the expression levels of ZO-1, claudin-5, and occludin were decreased, and brain water content and BBB permeability increased in the ICH model. The expression of CypA, NF-κB, and MMP-9 decreased significantly around the hematoma, while the expression of tight junction-related proteins increased significantly in response to 6KApoEp, especially at the 100 μg/kg dose. LRP expression increased around the ICH focus in response to 6KApoEp treatment, thus increasing the influence on the expression of CypA, NF-κB, and MMP-9. We conclude that 6KApoEp inhibits the CypA/NF-κB/MMP-9 pathway by activating LRP1, resulting in reduced BBB damage and less brain edema around the ICH. These results provide the theoretical basis for improving the prognosis and treatment of ICH. Our results suggest that 6KApoEp activates LRP1, resulting in the attenuation of tight junction protein degradation (ZO-1, occludin, and claudin-5) via the CypA/NF-κB/MMP-9 signaling pathway. The increased tight junction protein levels improve the BBB and attenuate edema development in brain tissue around the hematoma following ICH.
Collapse
Affiliation(s)
- Xing Chen
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China; Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Long Wang
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Ningning Wang
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Chen Li
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Hang Hang
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Guofeng Wu
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Siyin Ren
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Tan Jun
- Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Likun Wang
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China.
| |
Collapse
|
4
|
Liu J, Zhao W, Kang J, Li X, Han L, Hu Z, Zhou J, Meng X, Gao X, Zhang Y, Gu Y, Liu X, Chen X. Halcinonide activates smoothened to ameliorate ischemic stroke injury. Life Sci 2024:123324. [PMID: 39710062 DOI: 10.1016/j.lfs.2024.123324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
OBJECTIVES The Shh pathway may shed new light on developing new cell death inhibitors for the therapy of ischemic stroke. We aimed to examine whether the Shh co-reporter SMO or its agonist halcinonide can upregulate Bcl-2 to suppress neuronal cell death, ultimately improving behavioral deficits and reducing cerebral infarction in an ischemic stroke model. METHODS Halcinonide or genetic manipulation of SMO was conducted in PC12 cells to examine their impacts on oxidative or OGD/R stress, and the chemical, along with AAV-SMO or AAV-EGFP were tested in MCAO rats to investigate their potential protective effects against neuronal damages due to cerebral I/R injury. The amounts or activities of L-LA, LDH, ROS, MDA, SOD, MPO, GSSG, and GSH were detected using the corresponding biochemical kits. The levels of TNF-α and IL-6 were analyzed by ELISA. RESULTS The results show that halcinonide alleviated neurological score and cerebral infarction, and the abnormal changes in L-LA, LDH, MDA, SOD, MPO, GSH, GSSG, TNF-α, and IL-6 were also reversed in MCAO rats. Through expression or knockout of SMO, we discovered that SMO worked similarly to halcinonide, protecting neuronal cells from oxidative or OGD/R stress, and AAV-SMO prevented cerebral damages of MCAO rats caused by ischemia and reperfusion. Halcinonide inhibited Bcl-2/Bax-mediated apoptosis, at least partially by promoting the Shh signaling pathway through enhancing SMO expression in vivo and in vitro. CONCLUSION This study identified a new target and a candidate chemical for therapy of ischemic stroke, hopefully reducing its morbidity and mortality.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; School of Basic Medical Sciences, University of South China, Hengyang, Hunan 421001, PR China.
| | - Wenyang Zhao
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Jia Kang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Xiangxiang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Liang Han
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Zhuozhou Hu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Jing Zhou
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Xinrui Meng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Xiaoshan Gao
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Yixuan Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Youquan Gu
- Department of Neurology, First Hospital of Lanzhou University, Lanzhou 730000, PR China.
| | - Xiaohua Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China.
| | - Xinping Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
5
|
Daneshpour A, Shaka Z, Rezaei N. Interplay of cell death pathways and immune responses in ischemic stroke: insights into novel biomarkers. Rev Neurosci 2024:revneuro-2024-0128. [PMID: 39681004 DOI: 10.1515/revneuro-2024-0128] [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: 09/17/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024]
Abstract
Stroke is a severe neurological disease and a major worldwide issue, mostly manifesting as ischemic stroke (IS). In order to create effective treatments for IS, it is imperative to fully understand the underlying pathologies, as the existing therapeutic choices are inadequate. Recent investigations have shown the complex relationships between several programmed cell death (PCD) pathways, including necroptosis, ferroptosis, and pyroptosis, and their correlation with immune responses during IS. However, this relationship is still unclear. To address this gap, this review study explored the cellular interactions in the immune microenvironment of IS. Then, to validate prior findings and uncover biomarkers, the study investigated bioinformatics studies. Several pathways, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Toll-like receptor 4 (TLR4), and receptor-interacting protein kinase (RIPK), were involved in PCD-immune interactions. The bioinformatics studies reported key biomarkers such as glutathione peroxidase 4 (GPX4), NOD-like receptor family pyrin domain containing 3 (NLRP3), gasdermin D (GSDMD), and TLR4, which have important implications in ferroptosis, cuproptosis, pyroptosis, and necroptosis respectively. These biomarkers were associated with PCD mechanisms such as oxidative stress and inflammatory reactions. The immune infiltration analysis consistently revealed a significant correlation between PCD pathways and detrimental immune cells, such as neutrophils and γδ T cells. Conversely, M2 macrophages and T helper cells showed protective effects. In conclusion, considering the intricate network of interactions between immune responses and PCD pathways, this study emphasized the necessity of a paradigm shift in therapeutic approaches to address the injuries that are related to this complex network.
Collapse
Affiliation(s)
- Arian Daneshpour
- Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Zoha Shaka
- Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793 Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793 Iran
| |
Collapse
|
6
|
Tang X, Deng P, Li L, He Y, Wang J, Hao D, Yang H. Advances in genetically modified neural stem cell therapy for central nervous system injury and neurological diseases. Stem Cell Res Ther 2024; 15:482. [PMID: 39696712 DOI: 10.1186/s13287-024-04089-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Neural stem cells (NSCs) have increasingly been recognized as the most promising candidates for cell-based therapies for the central nervous system (CNS) injuries, primarily due to their pluripotent differentiation capabilities, as well as their remarkable secretory and homing properties. In recent years, extensive research efforts have been initiated to explore the therapeutic potential of NSC transplantation for CNS injuries, yielding significant advancements. Nevertheless, owing to the formation of adverse microenvironment at post-injury leading to suboptimal survival, differentiation, and integration within the host neural network of transplanted NSCs, NSC-based transplantation therapies often fall short of achieving optimal therapeutic outcomes. To address this challenge, genetic modification has been developed an attractive strategy to improve the outcomes of NSC therapies. This is mainly attributed to its potential to not only enhance the differentiation capacity of NSCs but also to boost a range of biological activities, such as the secretion of bioactive factors, anti-inflammatory effects, anti-apoptotic properties, immunomodulation, antioxidative functions, and angiogenesis. Furthermore, genetic modification empowers NSCs to play a more robust neuroprotective role in the context of nerve injury. In this review, we will provide an overview of recent advances in the roles and mechanisms of NSCs genetically modified with various therapeutic genes in the treatment of neural injuries and neural disorders. Also, an update on current technical parameters suitable for NSC transplantation and functional recovery in clinical studies are summarized.
Collapse
Affiliation(s)
- Xiangwen Tang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Peng Deng
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
- Basic Medical School Academy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Lin Li
- Basic Medical School Academy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuqing He
- Basic Medical School Academy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jinchao Wang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Dingjun Hao
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
7
|
Pfaff SJ, O'Reilly T, Zhang Y, Olsen W, Kuchenbecker K. Scp776, A Novel IGF-1 Fusion Protein for Acute Therapy to Promote Escape From Apoptosis in Tissues Affected by Ischemic Injury: 2 Randomized Placebo-Controlled Phase 1 Studies in Healthy Adults. Clin Pharmacol Drug Dev 2024. [PMID: 39648632 DOI: 10.1002/cpdd.1486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/23/2024] [Indexed: 12/10/2024]
Abstract
Apoptosis is a major driver of cell loss and infarct expansion in ischemic injuries such as acute ischemic stroke (AIS) and acute myocardial infarction (AMI). Insulin-like growth factor-1 (IGF-1) can mitigate cell death and potentiate recovery following acute ischemic injury, but short half-life and nonspecificity limit its therapeutic potential. Scp776 is an IGF-1 fusion protein designed to target damaged tissue and promote apoptosis escape and is in clinical development as an acute therapy for AIS and AMI. Two phase 1 placebo-controlled studies in healthy volunteers evaluated safety, tolerability, pharmacokinetic profile, and pharmacodynamics under single (1, 2, or 4 mg/kg) or multiple (6, 6.2, or 7.25 mg/kg total doses) dosing regimens. In addition, a blood glucose management plan was developed and implemented to mitigate hypoglycemia that may develop following scp776 injection. Scp776 was well tolerated in healthy volunteers (n = 51) without serious adverse events. Exposure increased in a near dose-proportional manner with a mean half-life across all doses of 8 hours. Adaptive dextrose infusions maintained normal blood glucose levels with occasional mild hypoglycemic events. These results informed scp776 dose selection and the design of blood glucose monitoring protocols for phase 2 studies.
Collapse
Affiliation(s)
- Samuel J Pfaff
- Silver Creek Pharmaceuticals, Inc., South San Francisco, California, USA
| | - Terry O'Reilly
- Silver Creek Pharmaceuticals, Inc., South San Francisco, California, USA
| | - Yan Zhang
- Silver Creek Pharmaceuticals, Inc., South San Francisco, California, USA
| | - Walter Olsen
- Silver Creek Pharmaceuticals, Inc., South San Francisco, California, USA
| | | |
Collapse
|
8
|
Gonzalez-Ortiz F, Holmegaard L, Andersson B, Brännmark C, Blomstrand C, Zetterberg H, Jood K, Blennow K, Jern C, Stanne TM. Plasma brain-derived tau correlates with cerebral infarct volume. J Intern Med 2024. [PMID: 39639627 DOI: 10.1111/joim.20041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND A blood-based biomarker that accurately reflects neuronal injury in acute ischemic stroke could be an easily accessible and cost-effective complement to clinical and radiological evaluation. Here, we investigate whether plasma levels of the novel biomarker brain-derived tau (BD-tau) reflect cerebral infarct volumes and whether BD-tau can improve clinical outcome prediction. METHODS The present study included 713 consecutive cases from two different hospital-based cohorts, the Sahlgrenska Academy Study on Ischemic Stroke (SAHLSIS) and SAHLSIS phase 2 (SAHLSIS2). Acute stroke severity was determined by the Scandinavian Stroke Scale converted to the National Institutes of Health stroke scale (NIHSS) in SAHLSIS and by the NIHSS in SAHLSIS2. All participants were assessed for functional outcome 3 months after stroke by the modified Rankin Scale, and 254 participants in SAHLSIS had quantitative neuroimaging available. FINDINGS Plasma BD-tau concentrations and cerebral infarct volumes were highly correlated (ρ 0.72, p < 0.001). BD-tau improved the prognostic accuracy of suffering an unfavorable outcome over age and stroke severity in the whole cohort. However, the gain in predictive power was dependent on stroke severity and infarct location. The largest improvement was observed for mild ischemic strokes (NIHSS <5; area under the curve [AUC] = 0.73 for age + NIHSS versus AUC = 0.84 with addition of BD-tau; DeLong p 0.02), posterior circulation stroke (AUC = 0.75 vs. AUC = 0.84; DeLong p 0.06) and more specifically for infarcts in the brainstem/cerebellum (AUC = 0.74 vs. 0.87; DeLong p 0.009). CONCLUSION Plasma BD-tau can provide information on the extent of acute neuronal damage in ischemic stroke and adds prognostic value for outcome, especially for mild and posterior circulation strokes.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lukas Holmegaard
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Björn Andersson
- Bioinformatics and Data Center, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Brännmark
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Medicine, Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Gothenburg, Sweden
| | - Christian Blomstrand
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Katarina Jood
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, China
| | - Christina Jern
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tara M Stanne
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
9
|
Ma XY, Qi CY, Xu XY, Li H, Liu CD, Wen XR, Fu YY, Liu Y, Liang J, Huang CY, Li DD, Li Y, Shen QC, Qi QZ, Zhu G, Wang N, Zhou XY, Song YJ. PRDX1 Interfering Peptide Disrupts Amino Acids 70-90 of PRDX1 to Inhibit the TLR4/NF-κB Signaling Pathway and Attenuate Neuroinflammation and Ischemic Brain Injury. Mol Neurobiol 2024; 61:10705-10721. [PMID: 38780721 DOI: 10.1007/s12035-024-04247-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Ischemic stroke ranks among the leading causes of death and disability in humans and is accompanied by motor and cognitive impairment. However, the precise mechanisms underlying injury after stroke and effective treatment strategies require further investigation. Peroxiredoxin-1 (PRDX1) triggers an extensive inflammatory cascade that plays a pivotal role in the pathology of ischemic stroke, resulting in severe brain damage from activated microglia. In the present study, we used molecular dynamics simulation and nuclear magnetic resonance to detect the interaction between PRDX1 and a specific interfering peptide. We used behavioral, morphological, and molecular experimental methods to demonstrate the effect of PRDX1-peptide on cerebral ischemia-reperfusion (I/R) in mice and to investigate the related mechanism. We found that PRDX1-peptide bound specifically to PRDX1 and improved motor and cognitive functions in I/R mice. In addition, pretreatment with PRDX1-peptide reduced the infarct area and decreased the number of apoptotic cells in the penumbra. Furthermore, PRDX1-peptide inhibited microglial activation and downregulated proinflammatory cytokines including IL-1β, IL-6, and TNF-α through inhibition of the TLR4/NF-κB signaling pathway, thereby attenuating ischemic brain injury. Our findings clarify the precise mechanism underlying PRDX1-induced inflammation after ischemic stroke and suggest that the PRDX1-peptide can significantly alleviate the postischemic inflammatory response by interfering with PRDX1 amino acids 70-90 and thereby inhibiting the TLR4/NF-κB signaling pathway. Our study provides a theoretical basis for a new therapeutic strategy to treat ischemic stroke.
Collapse
Affiliation(s)
- Xiang-Yu Ma
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Cheng-Yu Qi
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xing-Yi Xu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hui Li
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Chang-Dong Liu
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, 00000, Hong Kong SAR, China
| | - Xiang-Ru Wen
- Department of Chemistry, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yan-Yan Fu
- Department of Cell Biology and Neurobiology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yan Liu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jia Liang
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Cheng-Yu Huang
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Dan-Dan Li
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yan Li
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qian-Cheng Shen
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qian-Zhi Qi
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Guang Zhu
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, 00000, Hong Kong SAR, China
| | - Nan Wang
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiao-Yan Zhou
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Yuan-Jian Song
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China.
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
10
|
Song D, Li M, Zhang L, Zhang K, An Y, Feng M, Wang F, Yeh CT, Wang J, Guo F. Sphingosine-1-phosphate receptor 3 promotes neuronal apoptosis via the TNF-α/caspase-3 signaling pathway after acute intracerebral hemorrhage. Mol Cell Neurosci 2024; 131:103979. [PMID: 39581391 DOI: 10.1016/j.mcn.2024.103979] [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: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) has a high incidence and mortality rate among cerebrovascular diseases, and effective treatments are lacking. Sphingosine-1-phosphate receptor 3 (S1PR3) is associated with secondary immune inflammatory injury following ICH. However, its relationship with neuronal apoptosis and the specific underlying mechanism are not clear. METHODS We observed the effect of S1PR3 on neuronal apoptosis by assessing neurobehavioral scores, performing Western blot (WB) analysis, and performing TUNEL staining in a mouse model of ICH. Moreover, WBs and flow cytometry were used to study the specific mechanism and signaling pathways in HT22 cells in vitro. RESULTS The expression of S1PR3, CCL2, TNF-α, and cleaved-caspase-3 (c-caspase-3) and neuronal apoptosis were significantly increased after ICH, accompanied by neurobehavioral deterioration. These effects were significantly improved by treatment with CAY10444, a specific S1PR3 antagonist. After S1P stimulation of HT22 cells, the expression of S1PR3, CCL2, TNF-α and c-caspase-3 increased, and neuronal apoptosis increased by activating caspase-3 through the downstream PI3K/AKT apoptosis signaling pathway. After CAY10444 treatment, the expression of CCL2, TNF-α and c-caspase-3 was significantly reduced, and the PI3K/AKT apoptotic signaling pathway was regulated to reduce neuronal apoptosis. CONCLUSION An increase in S1P/S1PR3 after ICH may induce neuronal apoptosis by increasing TNF-α expression and activating the PI3K/AKT signaling pathway and the expression of caspase-3 effector proteins. CAY10444 can reduce neuronal apoptosis, improve symptoms and play a neuroprotective role by antagonizing S1PR3. S1PR3 may be a promising therapeutic target.
Collapse
Affiliation(s)
- Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Longxiao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaiyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan An
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengzhao Feng
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University Shuang-Ho Hospital, Taipei, Taiwan.
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
11
|
Guo Y, Mao H, Chen K, Dou W, Wang X. Impaired iron metabolism and cerebral perfusion patterns in unilateral middle cerebral artery stenosis or occlusion: Insights from quantitative susceptibility mapping. J Neuroradiol 2024; 52:101233. [PMID: 39547329 DOI: 10.1016/j.neurad.2024.101233] [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: 10/13/2023] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND AND PURPOSE Cerebral hypoperfusion caused by stenosis or occlusion of the middle cerebral artery (MCA) may be followed by impaired iron metabolism. We explored the association between iron changes of gray matter (GM) nuclei subregions and different cerebral perfusion patterns in patients with chronic unilateral middle cerebral artery (MCA) stenosis or occlusion using quantitative susceptibility imaging (QSM). METHODS Sixty-one patients with unilateral MCA stenosis or occlusion were recruited and scored with Alberta-Stroke-Program-Early-CT-Score (ASPECTS) based on relative cerebral blood flow (rCBF) measurements to calculate the number of corresponding hypoperfusion subregions, and then divided into an extensive-hypoperfusion group (EH group), regional-hypoperfusion group (RH group), and normal-perfusion group (Control group) accordingly. The measured magnetic susceptibility of GM nuclei subregions was compared between the lesion and contralateral side for each group and among the three groups. Correlation analysis was performed to assess the relationships of magnetic susceptibility of GM nuclei with mean rCBF, National-Institutes-of-Health-stroke-scale (NIHSS) and modified-Rankin-scale (mRS) scores. RESULTS Magnetic susceptibility in the putamen (PU) and globus pallidus (GP) at the lesion side was higher in the EH and RH groups compared with the contralateral side (all P < 0.05). Susceptibility in the lesion side PU and GP showed negative correlations with mean rCBF and positive correlations with NIHSS and mRS scores (all P < 0.05). CONCLUSION Our findings demonstrate that chronic cerebral hypoperfusion might be one cause of cerebral abnormal iron metabolism. In addition, magnetic susceptibility of PU and GP seems to be correlated with stroke scale scores, suggesting that iron deposition may play an important role in neurologic deficits after ischemic stroke.
Collapse
Affiliation(s)
- Yu Guo
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No.16766, Jingshi Rd, Jinan 250014, Shandong Province, PR China
| | - Huimin Mao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Kunjian Chen
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No.16766, Jingshi Rd, Jinan 250014, Shandong Province, PR China
| | - Weiqiang Dou
- MR Research, GE Healthcare, Beijing 10076, PR China.
| | - Xinyi Wang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No.16766, Jingshi Rd, Jinan 250014, Shandong Province, PR China.
| |
Collapse
|
12
|
Chen H, Wang M, Yang L, Li J, Li Z. Association of Uric Acid, High-Sensitivity C-Reactive Protein, and 90-Day Risk of Poor Function Outcome in Patients with Ischemic Stroke or Transient Ischemic Attacks. J Inflamm Res 2024; 17:8681-8694. [PMID: 39553311 PMCID: PMC11566209 DOI: 10.2147/jir.s494487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Aim The interaction between inflammatory biomarkers (high-sensitivity C-reactive protein, hsCRP) and antioxidants (uric acid, UA) regarding prognosis after ischemic stroke or transient ischemic attack (TIA) remains inadequately explored. This study aimed to assess (1) the individual and joint effects of hsCRP and UA, and (2) the neuroprotective role of UA in patients with elevated hsCRP levels concerning poor functional outcomes at 90 days. Methods A prospective cohort study was conducted involving 2140 consecutive ischemic stroke or TIA patients with hsCRP and UA levels. The primary outcome was defined as a poor functional outcome, indicated by a modified Rankin Scale (mRS) score of 3-6 at 90 days, with a shift in the mRS score as a secondary outcome. Logistic regression and propensity score (PS) analyses were employed to ensure robustness. Results Poor functional outcome occurred in 345 (16.1%) patients. Individual effects found that the highest quartiles of hsCRP (adjusted OR = 3.090; 95% CI 2.150-4.442) and UA (adjusted OR = 0.671; 95% CI 0.551-0.883) were associated with increased or decreased risk of poor functional outcome, respectively. Joint effects (adjusted OR = 3.994; 95% CI 2.758-5.640) between hsCRP and UA on the primary outcome were more apparent in patients with high hsCRP levels (hsCRP > 1.60 mg/L) and low UA levels (UA ≤ 291.85 µmol/L). For the patients with high hsCRP levels, patients with low UA levels had a higher risk of primary and secondary outcomes, compared with those with high UA levels, after unadjusted or adjusted for hsCRP. Similar and reliable results were observed in PS-based models. Conclusion In patients with ischemic stroke or TIA, joint high levels of hsCRP and low UA levels significantly correlate with increased risk of poor functional outcome at 90 days. In addition, high UA levels could reduce the risk of poor functional outcome for patients with high hsCRP levels.
Collapse
Affiliation(s)
- Haoran Chen
- Institute of Medical Information/Library, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Meng Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
- China National Clinical Research Center for Neurological Diseases, Beijing, People’s Republic of China
| | - Lin Yang
- Institute of Medical Information/Library, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Medical Information Intelligent Technology, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Jiao Li
- Institute of Medical Information/Library, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Medical Information Intelligent Technology, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
- China National Clinical Research Center for Neurological Diseases, Beijing, People’s Republic of China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
13
|
Wan Y, Xie Q, Hua Y, Xi G, Keep RF, Pandey A. Comparing white and gray matter responses to lobar intracerebral hemorrhage in piglets and the effects of deferoxamine. Exp Neurol 2024; 383:115041. [PMID: 39489370 DOI: 10.1016/j.expneurol.2024.115041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) often impacts patient white matter. However, preclinically, the effects of ICH are mostly studied in rodents with sparse white matter. This study used a lobar porcine ICH model to examine differences in the effects of ICH on white and gray matter as well as the role of the iron chelator deferoxamine (DFX), on attenuation of such injury. METHODS This two-part study was performed in piglets. Firstly, piglets had a needle (Sham) or 2.5 ml blood injection (ICH) and were euthanized at day 3. Secondly, animals were treated with vehicle or DFX after ICH and were euthanized at day 3. White and gray matter edema, the number of oligodendrocytes (mature and immature) and neurons, and the number of Perls' (iron), ferritin and heme oxygenase (HO)-1 positive cells were examined. RESULTS At day 3, ICH induced greater edema formation in white than gray matter. This marked white matter edema was associated with a loss of mature, but not immature, oligodendrocytes. ICH also induced neuronal death in gray matter. There were also marked increases in Perls', ferritin and HO-1 positive cells after ICH in both white and gray matter, but significantly more in the former. DFX attenuated ICH-induced brain edema in white but not gray matter and this was associated with increased survival of mature oligodendrocytes. DFX also increased survival of neurons in the gray matter and it reduced the number of Perls', ferritin and HO-1 positive cells in both tissue types. CONCLUSIONS While there were commonalities in perihematomal changes between white and gray matter after ICH, there was greater edema in white matter which may be linked to the susceptibility of mature oligodendrocytes to ICH injury. Similarly, while DFX reduced perihematomal iron overload in both white and gray matter, it only significantly reduced edema in white matter where it increased the number of mature oligodendrocytes.
Collapse
Affiliation(s)
- Yingfeng Wan
- Departments of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Qing Xie
- Departments of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Departments of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Departments of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Departments of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| | - Aditya Pandey
- Departments of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Goumtsa AF, Nguelefack-Mbuyo EP, Fofie CK, Fokoua AR, Becker A, Nguelefack TB. Neuroprotective effects of Aframomum pruinosum seed extract against stroke in rat: Role of antioxidant and anti-inflammatory mechanisms. J Stroke Cerebrovasc Dis 2024; 33:107942. [PMID: 39151813 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Stroke is a major cause of disability and neurological impairment worldwide. Effective prevention and management strategies are needed to reduce its burden. This study aimed to investigate the therapeutic effect of the seed ethanolic extract of Aframomum pruinosum (EEAP) on stroke and its related motor and cognitive deficits in rats. MATERIALS AND METHODS Stroke was induced by either middle cerebral artery occlusion (MCAO) or bilateral common carotid artery occlusion (BCCAO). In the MCAO model, rats received EEAP (75, 150, or 300 mg kg-1) or N-acetyl-L-cysteine (100 mg kg-1) orally for one week before 2 h of occlusion, followed by reperfusion. Twenty-four hours after ischemia, brain was collected for infarct size using 2, 3, 5 -TriphenylTetrazolium Chloride (TTC) staining, oxidative stress markers and inflammatory cytokines (TNF-α, IL-1β) measurements. In the BCCAO model, rats underwent occlusion for 30 min and received EEAP or quercetin (25 mg kg-1) for 7 days post-induction. Behavioral parameters were evaluated at the end of the treatment. Oxidative stress and inflammatory markers were measured in the cerebrum and cerebellum. RESULTS MCAO caused significant brain infarction, and increased lipid peroxidation, TNF-α and IL-1β contents. EEAP, rich in nerolidol, prevented these changes in a dose-dependent manner. BCCAO impaired the neurological function, mobility, and muscle strength of rats. It also increased lipid peroxidation and inflammatory cytokines in the cerebellum. EEAP significantly ameliorated these impairments. CONCLUSION EEAP exerts preventive and curative neuroprotective effects against ischemic stroke and its associated motor impairments at least partially through its antioxidant and anti-inflammatory properties, and its nerolidol content.
Collapse
Affiliation(s)
- Ariane Falone Goumtsa
- Research Unit of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67 Dschang, Cameroon
| | - Elvine Pami Nguelefack-Mbuyo
- Research Unit of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67 Dschang, Cameroon
| | - Christian Kuete Fofie
- Research Unit of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67 Dschang, Cameroon
| | - Aliance Romain Fokoua
- Research Unit of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67 Dschang, Cameroon
| | - Axel Becker
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Télesphore Benoît Nguelefack
- Research Unit of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67 Dschang, Cameroon.
| |
Collapse
|
15
|
Zhang X, Su G, Shao Z, Chan HW, Li S, Chow S, Tsang CK, Chow SF. Rational development of fingolimod nano-embedded microparticles as nose-to-brain neuroprotective therapy for ischemic stroke. Drug Deliv Transl Res 2024:10.1007/s13346-024-01721-8. [PMID: 39485637 DOI: 10.1007/s13346-024-01721-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
Ischemic stroke is one of the major diseases causing varying degrees of dysfunction and disability worldwide. The current management of ischemic stroke poses significant challenges due to short therapeutic windows and limited efficacy, highlighting the pressing need for novel neuroprotective treatment strategies. Previous studies have shown that fingolimod (FIN) is a promising neuroprotective drug. Here, we report the rational development of FIN nano-embedded nasal powders using full factorial design experiments, aiming to provide rapid neuroprotection after ischemic stroke. Flash nanoprecipitation was employed to produce FIN nanosuspensions with the aid of polyvinylpyrrolidone and cholesterol as stabilizers. The optimized nanosuspension (particle size = 134.0 ± 0.6 nm, PDI = 0.179 ± 0.021, physical stability = 72 ± 0 h, and encapsulation efficiency of FIN = 90.67 ± 0.08%) was subsequently spray-dried into a dry powder, which exhibited excellent redispersibility (RdI = 1.09 ± 0.04) and satisfactory drug deposition in the olfactory region using a customized 3D-printed nasal cast (45.4%) and an Alberta Idealized Nasal Inlet model (8.6%) at 15 L/min. The safety of the optimized FIN nano-embedded dry powder was confirmed in cytotoxicity studies with nasal (RPMI 2650 and Calu-3 cells) and brain related cells (SH-SY5Y and PC 12 cells), while the neuroprotective effects were demonstrated by observed behavioral improvements and reduced cerebral infarct size in a middle cerebral artery occlusion mouse stroke model. The neuroprotective effect was further evidenced by increased expression of anti-apoptotic protein BCL-2 and decreased expression of pro-apoptotic proteins CC3 and BAX in brain peri-infarct tissues. Our findings highlight the potential of nasal delivery of FIN nano-embedded dry powder as a rapid neuroprotective treatment strategy for acute ischemic stroke.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, L2-08B, 2/F, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Guangpu Su
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zitong Shao
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, L2-08B, 2/F, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Ho Wan Chan
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, L2-08B, 2/F, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Si Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, L2-08B, 2/F, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Stephanie Chow
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, L2-08B, 2/F, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Chi Kwan Tsang
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shing Fung Chow
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, L2-08B, 2/F, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong SAR, China.
| |
Collapse
|
16
|
Luwen H, Lei X, Qing-Rong O, Linlin L, Ming Y. Association between hs-CRP/HDL-C ratio and three-month unfavorable outcomes in patients with acute ischemic stroke: a second analysis based on a prospective cohort study. BMC Neurol 2024; 24:418. [PMID: 39468509 PMCID: PMC11514845 DOI: 10.1186/s12883-024-03929-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
OBJECTIVE The associations between the ratio of blood high-sensitivity C-reactive protein (hs-CRP) to high-density lipoprotein cholesterol (HDL-C) (hs-CRP/HDL-C ratio) and outcomes in patients with acute ischemic stroke (AIS) have yet to be established. This study is the first to examine the relationship between the hs-CRP/HDL-C ratio and three-month unfavorable outcomes in patients with AIS. METHODS This secondary analysis utilized data from a prospective cohort study involving 1559 AIS patients recruited at a South Korean hospital between January 2010 and December 2016. We constructed a binary logistic regression model to explore the association between the hs-CRP/HDL-C ratio and unfavorable outcomes in patients with AIS. An attempt was made to use a generalized additive model (GAM) with smooth curve fitting to elucidate potential nonlinear interactions. Furthermore, inflection points were identified via a recursive method, and binary logistic regression models were developed for each side of these inflection points. Ultimately, a log-likelihood ratio test was used to identify the most appropriate model for explaining the connection between the hs-CRP/HDL-C ratio and unfavorable outcomes in patients with AIS. RESULTS The incidence of unfavorable outcomes was 24.5%, with a median hs-CRP/HDL-C ratio of 3.64. After accounting for other factors, the binary logistic regression model revealed a statistically significant positive association between the hs-CRP/HDL-C ratio and the likelihood of poor outcomes in AIS patients (OR = 1.013, 95% CI: 1.005-1.022; P = 0.002). A nonlinear relationship was observed, with the first inflection point of the hs-CRP/HDL-C ratio at 42.74. Each 1-unit increase in the hs-CRP/HDL-C ratio was associated with a 2.4% greater risk of unfavorable outcomes (OR = 1.024, 95% CI: 1.011-1.038, P < 0.001). CONCLUSION This study provides evidence of a positive and nonlinear correlation between the hs-CRP/HDL-C ratio and poor three-month functional outcomes in AIS patients. When the hs-CRP/HDL-C ratio was less than 42.74, a positive association was observed with the risk of unfavorable outcomes. This finding offers a reference for optimizing early individualized therapy and aids in clinical counseling for patients with AIS.
Collapse
Affiliation(s)
- Huang Luwen
- Department of Neurology, Suining Central Hospital, Suining, Sichuan Province, 629000, China
| | - Xu Lei
- Department of Neurology, Suining Central Hospital, Suining, Sichuan Province, 629000, China
| | - Ouyang Qing-Rong
- Department of Neurology, Suining Central Hospital, Suining, Sichuan Province, 629000, China
| | - Li Linlin
- Department of Neurology, Suining Central Hospital, Suining, Sichuan Province, 629000, China
| | - Yu Ming
- Department of Neurology, Suining Central Hospital, Suining, Sichuan Province, 629000, China.
| |
Collapse
|
17
|
Wu B, Zhou D, Mei Z. Targeting the neurovascular unit: Therapeutic potential of traditional Chinese medicine for the treatment of stroke. Heliyon 2024; 10:e38200. [PMID: 39386825 PMCID: PMC11462356 DOI: 10.1016/j.heliyon.2024.e38200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Stroke poses a significant global health challenge due to its elevated disability and mortality rates, particularly affecting developing nations like China. The neurovascular unit (NVU), a new concept encompassing neurons, brain microvascular endothelial cells, pericytes, astrocytes, microglia, and the extracellular matrix, has gained prominence in recent years. Traditional Chinese medicine (TCM), deeply rooted in Chinese history, employs a combination of acupuncture and herbal treatments, demonstrating significant efficacy across all stages of stroke, notably during recovery. The holistic approach of TCM aligns with the NVU's comprehensive view of treating stroke by addressing neurons, surrounding cells, and blood vessels collectively. This review examines the role of NVU in stroke and endeavors to elucidate the mechanisms through which traditional Chinese medicine exerts its anti-stroke effects within the NVU framework. The NVU contributes to neuroinflammation, immune infiltration, blood-brain barrier permeability, oxidative stress, and Ca2+ overload during stroke occurs. Additionally, TCM targeting the NVU facilitates nerve repair post-stroke through various pathways and approaches. Specific herbs, including panax notoginseng, ginseng, and borneol, alleviate brain injury by enhancing brain-derived neurotrophic factor expression and targeting astrocytes and microglia to yield anti-inflammatory and antioxidant effects. Acupuncture, another facet of TCM, promotes brain injury repair by augmenting cerebral blood flow and improving circulation. This exploration aims to assess the viability of stroke treatment by directing TCM interventions toward the NVU, thus paving the way for its broader clinical application.
Collapse
Affiliation(s)
- Bingxin Wu
- Hubei Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, Hubei, 430000, China
| | - Dabiao Zhou
- Hubei Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, Hubei, 430000, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| |
Collapse
|
18
|
Li Y, He L, Song H, Bao X, Niu S, Bai J, Ma J, Yuan R, Liu S, Guo J. Cordyceps: Alleviating ischemic cardiovascular and cerebrovascular injury - A comprehensive review. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118321. [PMID: 38735418 DOI: 10.1016/j.jep.2024.118321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cordyceps has a long medicinal history as a nourishing herb in traditional Chinese medicine (TCM). Ischemic cardio-cerebrovascular diseases (CCVDs), including cerebral ischemic/reperfusion injury (CI/RI) and myocardial ischemic/reperfusion injury (MI/RI), are major contributors to mortality and disability in humans. Numerous studies have indicated that Cordyceps or its artificial substitutes have significant bioactivity on ischemic CCVDs, however, there is a lack of relevant reviews. AIM OF THE STUDY This review was conducted to investigate the chemical elements, pharmacological effects, clinical application and drug safety of Cordycepson ischemic CCVDs. MATERIALS AND METHODS A comprehensive search was conducted on the Web of Science, PubMed, Chinese National Knowledge Infrastructure (CNKI), and Wanfang databases using the keywords "Cordyceps", "Cerebral ischemic/reperfusion injury", and "Myocardial ischemic/reperfusion injury" or their synonyms. The retrieved literature was then categorized and summarized. RESULTS The study findings indicated that Cordyceps and its bioactive components, including adenosine, cordycepin, mannitol, polysaccharide, and protein, have the potential to protect against CI/RI and MI/RI by improving blood perfusion, mitigating damage from reactive oxygen species, suppressing inflammation, preventing cellular apoptosis, and promoting tissue regeneration. Individually, Cordyceps could reduce neuronal excitatory toxicity and blood-brain barrier damage caused by cerebral ischemia. It can also significantly improve cardiac energy metabolism disorders and inhibit calcium overload caused by myocardial ischemia. Additionally, Cordyceps exerts a significant preventive or curative influence on the factors responsible for heart/brain ischemia, including hypertension, thrombosis, atherosclerosis, and arrhythmia. CONCLUSION This study demonstrates Cordyceps' prospective efficacy and safety in the prevention or treatment of CI/RI and MI/RI, providing novel insights for managing ischemic CCVDs.
Collapse
Affiliation(s)
- Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Liying He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Haoran Song
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Xiuwen Bao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Shuqi Niu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Jing Bai
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Junhao Ma
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Run Yuan
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Sijing Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Jinlin Guo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
19
|
Xu C, Jiang F, Mao Y, Wei W, Song J, Jia F, Du X, Zhong D, Li G. Disulfiram attenuates cell and tissue damage and blood‒brain barrier dysfunction after intracranial haemorrhage by inhibiting the classical pyroptosis pathway. Sci Rep 2024; 14:21860. [PMID: 39300102 DOI: 10.1038/s41598-024-67118-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/08/2024] [Indexed: 09/22/2024] Open
Abstract
No single treatment significantly reduces the mortality rate and improves neurological outcomes after intracerebral haemorrhage (ICH). New evidence suggests that pyroptosis-specific proteins are highly expressed in the perihaematomal tissues of patients with ICH and that the disulfiram (DSF) inhibits pyroptosis. An ICH model was established in C57BL/6 mice by intracranial injection of collagenase, after which DSF was used to treat the mice. Cell model of ICH was constructed, and DSF was used to treat the cells. HE, TUNEL, Nissl, FJC and IF staining were performed to evaluate the morphology of brain tissues; Western blotting and ELISA were performed to measure the protein expression of NOD-like receptor protein 3 (NLRP3)/Caspase-1/gasdermin D (GSDMD) classical pyroptosis pathway and Toll-likereceptor4 (TLR4)/nuclear factor-kappaB (NF-κB) inflammatory signaling pathway and blood‒brain barrier-associated factoes, and the wet/dry weight method was used to determine the brain water content. The expression of proteins related to the NLRP3/Caspase-1/GSDMD pathway and the TLR4/NF-κB pathway was upregulated in tissues surrounding the haematoma compared with that in control tissues; Moreover, the expression of the blood-brain barrier structural proteins occludin and zonula occludens-1 (ZO-1) was downregulated, and the expression of Aquaporin Protein-4 (AQP4) and matrix metalloprotein 9 (MMP-9) was upregulated. DSF significantly inhibited these changes, reduced the haematoma volume, decreased the brain water content, reduced neuronal death and degeneration and improved neurological function after ICH. ICH activated the classical pyroptosis pathway and TLR4/NF-κB inflammatory pathway, disruped the expression of blood-brain barrier structural proteins, and exacerbated brain injury and neurological dysfunction. DSF inhibited these changes and exerted the therapeutic effects on pathological changes and dysfunction caused by ICH.
Collapse
Affiliation(s)
- Chen Xu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangchao Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanfu Mao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan Wei
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihe Song
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feihong Jia
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinshu Du
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Liu R, Yu Y, Ge Q, Feng R, Zhong G, Luo L, Han Z, Wang T, Huang C, Xue J, Huang Z. Genistein-3'-sodium sulfonate promotes brain functional rehabilitation in ischemic stroke rats by regulating astrocytes polarization through NF-κB signaling pathway. Chem Biol Interact 2024; 400:111159. [PMID: 39059603 DOI: 10.1016/j.cbi.2024.111159] [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: 06/05/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
The activation and polarization of astrocytes are involved in neuroinflammation and brain functional rehabilitation after ischemic stroke. Our previous studies display the neuroprotective effect of genistein-3'-sodium sulfonate (GSS) in the acute phase of cerebral ischemia-reperfusion injury (CI/RI). This study aimed to investigate the brain function improvement of GSS during the recovery period after CI/RI in rats and to explore the potential mechanism from the perspective of astrocyte activation and polarization. The transient middle cerebral artery occlusion (tMCAO) rats were treated with GSS (1 mg/kg) continuously for 28 days. The behavior tests were measured to assess neurological function. The mRNA and protein expression in affected cerebral cortex were detected on day 29 after tMCAO. Our results demonstrated that GSS treatment significantly improved the spatial and temporal gait parameters in the Catwalk gait test, prolonged the time on the stick and increased the rotation speed in the rotarod test, and decreased the time to find the hidden platform and increased the time in the target quadrant in the Morris water maze test. In addition, GFAP, GBP2, C3, IL-1β protein expressions and Nos2A mRNA level were decreased, while Nrf2, BDNF, IL-10 protein expressions and Sphk1 and Nef2l2 mRNA levels increased after GSS treatment. Interestingly, GSS presented a strong binding affinity to TLR4 and suppressed the activation of NF-κB signaling. In conclusion, GSS can promote brain function recovery by inhibiting astrocyte activation and polarization to A1 phenotype, and enhancing astrocyte polarization to A2 phenotype via inactivating TLR4/NF-κB signaling, which provide a candidate compound for clinical rehabilitation therapy in the recovery period after ischemic stroke.
Collapse
Affiliation(s)
- Ruizhen Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China; School of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Yunling Yu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Qinglian Ge
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Ruixue Feng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Guixiang Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Li Luo
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Zun Han
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Tianyun Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Cheng Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jinhua Xue
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China.
| | - Zhihua Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China; School of Basic Medical Sciences, China Medical University, Shenyang 110122, China.
| |
Collapse
|
21
|
Brooks B, D'Egidio F, Borlongan MC, Borlongan MC, Lee JY. Stem cell grafts enhance endogenous extracellular vesicle expression in the stroke brain. Brain Res Bull 2024; 214:110999. [PMID: 38851436 DOI: 10.1016/j.brainresbull.2024.110999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Endogenous brain repair occurs following an ischemic stroke but is transient, thus unable to fully mount a neuroprotective response against the evolving secondary cell death. Finding a treatment strategy that may render robust and long-lasting therapeutic effects stands as a clinically relevant therapy for stroke. Extracellular vesicles appear to be upregulated after stroke, which may represent a candidate target for neuroprotection. In this study, we probed whether transplanted stem cells could enhance the expression of extracellular vesicles to afford stable tissue remodeling in the ischemic stroke brain. Aged rats were initially exposed to the established ischemic stroke model of middle cerebral artery occlusion then received intravenous delivery of either bone marrow-derived mesenchymal stem cell transplantation or vehicle. A year later, the animals were assayed for brain damage, inflammation, and extracellular vesicle expression. Our findings revealed that while core infarction was not reduced, the stroke animals transplanted with stem cells displayed a significant reduction in peri-infarct cell loss that coincided with downregulated Iba1-labeled inflammatory cells and upregulated CD63-positive extracellular vesicles that appeared to be co-localized with GFAP-positive astrocytes. Interestingly, grafted stem cells were not detected at one year post-transplantation period, suggesting that the extracellular vesicles likely originated within the host brain. That long-lasting functional benefits persisted in the absence of surviving transplanted stem cells, but with upregulation of endogenous extracellular vesicles, advances the concept that transplantation of stem cells acutely after stroke propels host extracellular vesicles to the ischemic brain, altogether promoting chronic brain remodeling.
Collapse
Affiliation(s)
- Beverly Brooks
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Francesco D'Egidio
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Maximillian C Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Mia C Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States.
| |
Collapse
|
22
|
Klepinowski T, Skonieczna-Żydecka K, Łoniewski I, Pettersson SD, Wierzbicka-Woś A, Kaczmarczyk M, Palma J, Sawicki M, Taterra D, Poncyljusz W, Alshafai NS, Stachowska E, Ogilvy CS, Sagan L. A prospective pilot study of gut microbiome in cerebral vasospasm and delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Sci Rep 2024; 14:17617. [PMID: 39080476 PMCID: PMC11289281 DOI: 10.1038/s41598-024-68722-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: 02/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
A recent systematic review indicated that gut-microbiota-brain axis contributes to growth and rupture of intracranial aneurysms. However, gaps were detected in the role of intestinal microbiome in cerebral vasospasm (CVS) after aneurysmal subarachnoid hemorrhage (aSAH). This is the first pilot study aiming to test study feasibility and identify differences in gut microbiota between subjects with and without CVS following aSAH. A prospective nested case-control pilot study with 1:1 matching was conducted recruiting subjects with aSAH: cases with CVS; and controls without CVS based on the clinical picture and structured bedside transcranial Doppler (TCD). Fecal samples for microbiota analyses by means of 16S rRNA gene amplicon sequencing were collected within the first 96 h after ictus. Operational taxonomic unit tables were constructed, diversity metrics calculated, phylogenetic trees built, and differential abundance analysis (DAA) performed. At baseline, the groups did not differ significantly in basic demographic and aneurysm-related characteristics (p > 0.05). Alpha-diversity (richness and Shannon Index) was significantly reduced in cases of middle cerebral artery (MCA) vasospasm (p < 0.05). In DAA, relative abundance of genus Acidaminococcus was associated with MCA vasospasm (p = 0.00013). Two butyrate-producing genera, Intestinimonas and Butyricimonas, as well as [Clostridium] innocuum group had the strongest negative correlation with the mean blood flow velocity in anterior cerebral arteries (p < 0.01; rho = - 0.63; - 0.57, and - 0.57, respectively). In total, 16 gut microbial genera were identified to correlate with TCD parameters, and two intestinal genera correlated with outcome upon discharge. In this pilot study, we prove study feasibility and present the first preliminary evidence of gut microbiome signature associating with CVS as a significant cause of stroke in subjects with aSAH.
Collapse
Affiliation(s)
- Tomasz Klepinowski
- Department of Neurosurgery, Pomeranian Medical University Hospital no. 1, Unii Lubelskiej 1, 71-252, Szczecin, Poland.
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland.
| | - Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
| | - Samuel D Pettersson
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Anna Wierzbicka-Woś
- Research and Development Centre, Sanprobi sp. z o.o. sp. K, Kurza Stopka 5/c, 70-535, Szczecin, Poland
| | - Mariusz Kaczmarczyk
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
- Research and Development Centre, Sanprobi sp. z o.o. sp. K, Kurza Stopka 5/c, 70-535, Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
| | - Marcin Sawicki
- Department of Diagnostic Imaging and Interventional Radiology, Pomeranian Medical University in Szczecin Hospital no. 1, Szczecin, Poland
| | - Dominik Taterra
- Department of Orthopedics and Rehabilitation, Jagiellonian University Medical College, Zakopane, Poland
| | - Wojciech Poncyljusz
- Department of Diagnostic Imaging and Interventional Radiology, Pomeranian Medical University in Szczecin Hospital no. 1, Szczecin, Poland
| | | | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Broniewskiego 24, 70-204, Szczecin, Poland
| | - Christopher S Ogilvy
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leszek Sagan
- Department of Neurosurgery, Pomeranian Medical University Hospital no. 1, Unii Lubelskiej 1, 71-252, Szczecin, Poland
| |
Collapse
|
23
|
Yang H, Ibrahim MM, Zhang S, Sun Y, Chang J, Qi H, Yang S. Targeting post-stroke neuroinflammation with Salvianolic acid A: molecular mechanisms and preclinical evidence. Front Immunol 2024; 15:1433590. [PMID: 39139557 PMCID: PMC11319147 DOI: 10.3389/fimmu.2024.1433590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Salvianolic acid A (SalA), a bioactive compound extracted from Salvia miltiorrhiza, has garnered considerable interest for its potential in ameliorating the post-stroke neuroinflammation. This review delineates the possible molecular underpinnings of anti-inflammatory and neuroprotective roles of SalA, offering a comprehensive analysis of its therapeutic efficacy in preclinical studies of ischemic stroke. We explore the intricate interplay between post-stroke neuroinflammation and the modulatory effects of SalA on pro-inflammatory cytokines, inflammatory signaling pathways, the peripheral immune cell infiltration through blood-brain barrier disruption, and endothelial cell function. The pharmacokinetic profiles of SalA in the context of stroke, characterized by enhanced cerebral penetration post-ischemia, makes it particularly suitable as a therapeutic agent. Preliminary clinical findings have demonstrated that salvianolic acids (SA) has a positive impact on cerebral perfusion and neurological deficits in stroke patients, warranting further investigation. This review emphasizes SalA as a potential anti-inflammatory agent for the advancement of innovative therapeutic approaches in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongchun Yang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Muhammad Mustapha Ibrahim
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Siyu Zhang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Sun
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Qi
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shilun Yang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
24
|
de Oliveira RMW, Kohara NA, Milani H. Cannabidiol in experimental cerebral ischemia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 177:95-120. [PMID: 39029992 DOI: 10.1016/bs.irn.2024.04.012] [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: 07/21/2024]
Abstract
The absence of blood flow in cerebral ischemic conditions triggers a multitude of intricate pathophysiological mechanisms, including excitotoxicity, oxidative stress, neuroinflammation, disruption of the blood-brain barrier and white matter disarrangement. Despite numerous experimental studies conducted in preclinical settings, existing treatments for cerebral ischemia (CI), such as mechanical and pharmacological therapies, remain constrained and often entail significant side effects. Therefore, there is an imperative to explore innovative strategies for addressing CI outcomes. Cannabidiol (CBD), the most abundant non-psychotomimetic compound derived from Cannabis sativa, is a pleiotropic substance that interacts with diverse molecular targets and has the potential to influence various pathophysiological processes, thereby contributing to enhanced outcomes in CI. This chapter provides a comprehensive overview of the primary effects of CBD in in vitro and diverse animal models of CI and delves into some of its plausible mechanisms of neuroprotection.
Collapse
Affiliation(s)
| | - Nathalia Akemi Kohara
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Paraná, Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
25
|
Liu H, Li H, Chen T, Yu F, Lin Q, Zhao H, Jin L, Peng R. Research Progress on Micro(nano)plastic-Induced Programmed Cell Death Associated with Disease Risks. TOXICS 2024; 12:493. [PMID: 39058145 PMCID: PMC11281249 DOI: 10.3390/toxics12070493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
Due to their robust migration capabilities, slow degradation, and propensity for adsorbing environmental pollutants, micro(nano)plastics (MNPs) are pervasive across diverse ecosystems. They infiltrate various organisms within different food chains through multiple pathways including inhalation and dermal contact, and pose a significant environmental challenge in the 21st century. Research indicates that MNPs pose health threats to a broad range of organisms, including humans. Currently, extensive detection data and studies using experimental animals and in vitro cell culture indicate that MNPs can trigger various forms of programmed cell death (PCD) and can induce various diseases. This review provides a comprehensive and systematic analysis of different MNP-induced PCD processes, including pyroptosis, ferroptosis, autophagy, necroptosis, and apoptosis, based on recent research findings and focuses on elucidating the links between PCD and diseases. Additionally, targeted therapeutic interventions for these diseases are described. This review provides original insights into the opportunities and challenges posed by current research findings. This review evaluates ways to mitigate various diseases resulting from cell death patterns. Moreover, this paper enhances the understanding of the biohazards associated with MNPs by providing a systematic reference for subsequent toxicological research and health risk mitigation efforts.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (H.L.); (H.L.); (T.C.); (F.Y.); (Q.L.); (H.Z.); (L.J.)
| |
Collapse
|
26
|
Li G, Zhao Y, Ma W, Gao Y, Zhao C. Systems-level computational modeling in ischemic stroke: from cells to patients. Front Physiol 2024; 15:1394740. [PMID: 39015225 PMCID: PMC11250596 DOI: 10.3389/fphys.2024.1394740] [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: 03/02/2024] [Accepted: 06/14/2024] [Indexed: 07/18/2024] Open
Abstract
Ischemic stroke, a significant threat to human life and health, refers to a class of conditions where brain tissue damage is induced following decreased cerebral blood flow. The incidence of ischemic stroke has been steadily increasing globally, and its disease mechanisms are highly complex and involve a multitude of biological mechanisms at various scales from genes all the way to the human body system that can affect the stroke onset, progression, treatment, and prognosis. To complement conventional experimental research methods, computational systems biology modeling can integrate and describe the pathogenic mechanisms of ischemic stroke across multiple biological scales and help identify emergent modulatory principles that drive disease progression and recovery. In addition, by running virtual experiments and trials in computers, these models can efficiently predict and evaluate outcomes of different treatment methods and thereby assist clinical decision-making. In this review, we summarize the current research and application of systems-level computational modeling in the field of ischemic stroke from the multiscale mechanism-based, physics-based and omics-based perspectives and discuss how modeling-driven research frameworks can deliver insights for future stroke research and drug development.
Collapse
Affiliation(s)
- Geli Li
- Gusu School, Nanjing Medical University, Suzhou, China
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yanyong Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Wen Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, Nanjing, China
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Pinosanu LR, Boboc IKS, Balseanu TA, Gresita A, Hermann DM, Popa-Wagner A, Catalin B. Beam narrowing test: a motor index of post-stroke motor evaluation in an aged rat model of cerebral ischemia. J Neural Transm (Vienna) 2024; 131:763-771. [PMID: 38598100 PMCID: PMC11199207 DOI: 10.1007/s00702-024-02768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
Each year, 15 million people worldwide suffer from strokes. Consequently, researchers face increasing pressure to develop reliable behavioural tests for assessing functional recovery after a stroke. Our aim was to establish a new motor performance index that can be used to evaluate post-stroke recovery in both young and aged animals. Furthermore, we validate the proposed procedure and recommend the necessary number of animals for experimental stroke studies. Young (n = 20) and aged (n = 27) Sprague-Dawley rats were randomly assigned to receive either sham or stroke surgery. The newly proposed performance index was calculated for the post-stroke acute, subacute and chronic phases. The advantage of using our test over current tests lies in the fact that the newly proposed motor index test evaluates not only the performance of the unaffected side in comparison to the affected one but also assesses overall performance by taking into account speed and coordination. Moreover, it reduces the number of animals needed to achieve a statistical power of 80%. This aspect is particularly crucial when studying aged rodents. Our approach can be used to monitor and assess the effectiveness of stroke therapies in experimental models using aged animals.
Collapse
Affiliation(s)
- Leonard Radu Pinosanu
- Department of Pharmacology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Ianis Kevyn Stefan Boboc
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
- Department of Pharmacology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Tudor Adrian Balseanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Andrei Gresita
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, 115680-8000, USA
| | - Dirk M Hermann
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Aurel Popa-Wagner
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Bogdan Catalin
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, 115680-8000, USA
| |
Collapse
|
28
|
Guo D, Hu L, Xie P, Sun P, Yu W. Seipin is involved in oxygen-glucose deprivation/reoxygenation induced neuroinflammation by regulating the TLR3/TRAF3/NF-κB pathway. Int Immunopharmacol 2024; 134:112182. [PMID: 38703568 DOI: 10.1016/j.intimp.2024.112182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Seipin plays a crucial role in lipid metabolism and is involved in neurological disorders. However, the function and mechanism of action of seipin in acute ischemic stroke have not yet been elucidated. Here, we aimed to investigate the effect of seipin on neuroinflammation induced by oxygen-glucose deprivation/reoxygenation (OGD/R) and further explore the molecular mechanism by functional experiments. Our results revealed a significant decrease in seipin mRNA levels, accompanied by enhanced expression of TNF-α in patients with AIS, and a significant negative correlation between seipin and TNF-α was observed. Additionally, there was a negative correlation between seipin levels and the National Institutes of Health Stroke Scale (NIHSS) score. Furthermore, seipin levels were also decreased in middle cerebral artery occlusion/reperfusion (MCAO/R) mice and OGD/R-treated BV2 cells. RNA sequencing analysis showed that seipin knockdown altered the Toll-like receptor 3 (TLR3) signaling pathway. It was further confirmed in vitro that seipin knockdown caused significantly increased secretion of inflammatory factors including TNF-α, interleukin (IL)-1β, and interferon (IFN)-β. Meanwhile, seipin knockdown activated the Tlr3 signal pathway while this effect could be reversed by Tlr3 inhibitor in OGD/R treated BV2 cells. Furthermore, neuroinflammation induced by OGD/R was significantly reduced by seipin overexpression. Overall, our study demonstrate that seipin deficiency aggravates neuroinflammation by activating the TLR3/TRAF3/NF-κB signaling pathway after OGD/R stimuli, and suggest that seipin may be a potential therapeutic target for AIS.
Collapse
Affiliation(s)
- Dongfen Guo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China
| | - Lele Hu
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550023, Guizhou, China
| | - Peng Xie
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China
| | - Ping Sun
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550023, Guizhou, China.
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China; Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang 550025, Guizhou, China.
| |
Collapse
|
29
|
Zhao Q, Chen L, Zhang X, Yang H, Li Y, Li P. β-elemene promotes microglial M2-like polarization against ischemic stroke via AKT/mTOR signaling axis-mediated autophagy. Chin Med 2024; 19:86. [PMID: 38879549 PMCID: PMC11179363 DOI: 10.1186/s13020-024-00946-6] [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: 03/26/2024] [Accepted: 05/14/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND Resident microglia- and peripheric macrophage-mediated neuroinflammation plays a predominant role in the occurrence and development of ischemic stroke. Microglia undergo polarization to M1/M2-like phenotype under stress stimulation, which mediates intracellular inflammatory response. β-elemene is a natural sesquiterpene and possesses potent anti-inflammatory activity. This study aimed to investigate the anti-inflammatory efficacy and mechanism of β-elemene in ischemic stroke from the perspective of balancing microglia M1/M2-like polarization. METHODS The middle cerebral artery occlusion (MCAO) model and photothrombotic stroke model were established to explore the regulation effect of β-elemene on the cerebral ischemic injury. The LPS and IFN-γ stimulated BV-2 cells were used to demonstrate the anti-inflammatory effects and potential mechanism of β-elemene regulating M1/M2-like polarization in vitro. RESULTS In C57BL/6 J mice subjected to MCAO model and photothrombotic stroke model, β-elemene attenuated neurological deficit, reduced the infarction volume and neuroinflammation, thus improving ischemic stroke injury. β-elemene promoted the phenotype transformation of microglia from M1-like to M2-like, which prevented neurons from oxygen and glucose deprivation/reoxygenation (OGD/R) injury by inhibiting inflammatory factor release, thereby reducing neuronal apoptosis. Mechanically, β-elemene prevented the activation of TLR4/NF-κΒ and MAPK signaling pathway and increased AKT/mTOR mediated-autophagy, thereby promoting M2-like polarization of microglia. CONCLUSIONS These results indicated that β-elemene improved cerebral ischemic injury and promoted the transformation of microglia phenotype from M1-like to M2-like, at least in part, through AKT/mTOR-mediated autophagy. This study demonstrated that β-elemene might serve as a promising drug for alleviating ischemic stroke injury.
Collapse
Affiliation(s)
- Qiong Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, 211198, China
| | - Lu Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, 211198, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, 211198, China
| | - Hua Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, 211198, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, 211198, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, 211198, China.
| |
Collapse
|
30
|
Maida CD, Norrito RL, Rizzica S, Mazzola M, Scarantino ER, Tuttolomondo A. Molecular Pathogenesis of Ischemic and Hemorrhagic Strokes: Background and Therapeutic Approaches. Int J Mol Sci 2024; 25:6297. [PMID: 38928006 PMCID: PMC11203482 DOI: 10.3390/ijms25126297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke represents one of the neurological diseases most responsible for death and permanent disability in the world. Different factors, such as thrombus, emboli and atherosclerosis, take part in the intricate pathophysiology of stroke. Comprehending the molecular processes involved in this mechanism is crucial to developing new, specific and efficient treatments. Some common mechanisms are excitotoxicity and calcium overload, oxidative stress and neuroinflammation. Furthermore, non-coding RNAs (ncRNAs) are critical in pathophysiology and recovery after cerebral ischemia. ncRNAs, particularly microRNAs, and long non-coding RNAs (lncRNAs) are essential for angiogenesis and neuroprotection, and they have been suggested to be therapeutic, diagnostic and prognostic tools in cerebrovascular diseases, including stroke. This review summarizes the intricate molecular mechanisms underlying ischemic and hemorrhagic stroke and delves into the function of miRNAs in the development of brain damage. Furthermore, we will analyze new perspectives on treatment based on molecular mechanisms in addition to traditional stroke therapies.
Collapse
Affiliation(s)
- Carlo Domenico Maida
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy;
- Molecular and Clinical Medicine Ph.D. Programme, University of Palermo, 90133 Palermo, Italy
| | - Rosario Luca Norrito
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| | - Salvatore Rizzica
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy;
| | - Marco Mazzola
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| | - Elisa Rita Scarantino
- Division of Geriatric and Intensive Care Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence, 50134 Florence, Italy;
| | - Antonino Tuttolomondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| |
Collapse
|
31
|
Hong L, Zhuo T, Jing S. Silencing of METTL3 inhibits m6A methylation of NEK7 to suppress pyrolysis in an HT-22 cell-based model of intracerebral hemorrhage. Brain Res 2024; 1831:148828. [PMID: 38408556 DOI: 10.1016/j.brainres.2024.148828] [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: 12/18/2023] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Intracerebral hemorrhage (ICH) induces severe neurological damage, and its progression is driven by METTL3. This study aimed to investigate the role of METTL3 in ICH via in vitro experiments. For this purpose, HT-22 cells were treated with hemin to mimic ICH in vitro, followed by evaluating cell pyroptosis using flow cytometry, lactic dehydrogenase release analysis, enzyme-linked immunosorbent assay, and western blotting. Moreover, N6-methyl adenosine (m6A) methylation of NEK7 was assessed using methylated RNA immunoprecipitation, RNA immunoprecipitation, dual-luciferase reporter assay, and quantitative real-time polymerase chain reaction. Results indicated that knockdown of METTL3 inhibited hemin-induced pyroptosis and suppressed m6A methylation of NEK7 due to METTL3 downregulation, reducing NEK7 mRNA stability. The effects on METTL3-induced cell pyroptosis were abrogated by overexpressing NEK7, while IGF2BP2 increased NEK7 expression. Similarly, IGF2BP2 silence downregulated NEK7 expression mediated by METTL3. In conclusion, silencing of METTL3 inhibited hemin-induced HT-22 cell pyroptosis by suppressing m6A methylation of NEK7, which was recognized by IGF2BP2. These findings are envisaged to identify a novel therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Lei Hong
- Department of Neurosurgery, Ningbo Ninth Hospital, Ningbo, Zhejiang Province 315000, China.
| | - Ting Zhuo
- Department of Neurosurgery, Ningbo Ninth Hospital, Ningbo, Zhejiang Province 315000, China
| | - Shuguang Jing
- Department of Neurosurgery, Ningbo Ninth Hospital, Ningbo, Zhejiang Province 315000, China
| |
Collapse
|
32
|
Farzaneh M, Anbiyaee O, Azizidoost S, Nasrolahi A, Ghaedrahmati F, Kempisty B, Mozdziak P, Khoshnam SE, Najafi S. The Mechanisms of Long Non-coding RNA-XIST in Ischemic Stroke: Insights into Functional Roles and Therapeutic Potential. Mol Neurobiol 2024; 61:2745-2753. [PMID: 37932544 DOI: 10.1007/s12035-023-03740-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023]
Abstract
Ischemic stroke, which occurs due to the occlusion of cerebral arteries, is a common type of stroke. Recent research has highlighted the important role of long non-coding RNAs (lncRNAs) in the development of cerebrovascular diseases, specifically ischemic stroke. Understanding the functional roles of lncRNAs in ischemic stroke is crucial, given their potential contribution to the disease pathology. One noteworthy lncRNA is X-inactive specific transcript (XIST), which exhibits downregulation during the early stages of ischemic stroke and subsequent upregulation in later stages. XIST exert its influence on the development of ischemic stroke through interactions with multiple miRNAs and transcription factors. These interactions play a significant role in the pathogenesis of the condition. In this review, we have provided a comprehensive summary of the functional roles of XIST in ischemic stroke. By investigating the involvement of XIST in the disease process, we aim to enhance our understanding of the mechanisms underlying ischemic stroke and potentially identify novel therapeutic targets.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, Namazi Hospital, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bartosz Kempisty
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wrocław, Poland
- Institute of Veterinary Medicine, Department of Veterinary Surgery, Nicolaus Copernicus University, Torun, Poland
- North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC, 27695, USA
| | - Paul Mozdziak
- North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC, 27695, USA
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Salman M, Stayton AS, Parveen K, Parveen A, Puchowicz MA, Parvez S, Bajwa A, Ishrat T. Intranasal Delivery of Mitochondria Attenuates Brain Injury by AMPK and SIRT1/PGC-1α Pathways in a Murine Model of Photothrombotic Stroke. Mol Neurobiol 2024; 61:2822-2838. [PMID: 37946007 DOI: 10.1007/s12035-023-03739-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Ischemic stroke is one of the major causes of morbidity and mortality worldwide. Mitochondria play a vital role in the pathological processes of cerebral ischemic injury, but its transplantation and underlying mechanisms remain unclear. In the present study, we examined the effects of mitochondrial therapy on the modulation of AMPK and SIRT1/PGC-1α signaling pathway, oxidative stress, and NLRP3 inflammasome activation after photothrombotic ischemic stroke (pt-MCAO). The adult male mice were subjected to the pt-MCAO in which the proximal-middle cerebral artery was exposed with a 532-nm laser beam for 4 min by retro-orbital injection of a photosensitive dye (Rose Bengal: 15 mg/kg) before the laser light exposure and isolated mitochondria (100 μg protein) were administered intranasally at 30 min, 24 h, and 48 h following post-stroke. After 72 h, mice were tested for neurobehavioral outcomes and euthanized for infarct volume, brain edema, and molecular analysis. First, we found that mitochondria therapy significantly decreased brain infarct volume and brain edema, improved neurological dysfunction, attenuated ischemic stroke-induced oxidative stress, and neuroinflammation. Second, mitochondria treatment inhibited NLRP3 inflammasome activation. Finally, mitochondria therapy accelerated p-AMPKα(Thr172) and PGC-1α expression and resorted SIRT1 protein expression levels in pt-MCAO mice. In conclusion, our results demonstrate that mitochondria therapy exerts neuroprotective effects by inhibiting oxidative damage and inflammation, mainly dependent on the heightening activation of the AMPK and SIRT1/PGC-1α signaling pathway. Thus, intranasal delivery of mitochondria might be considered a new therapeutic strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Mohd Salman
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-228, Memphis, TN, 38163, USA.
| | - Amanda S Stayton
- Transplant Research Institute, College of Medicine, The University of Tennessee Health Science Center, 71 S Manassas St, Room 418H, Memphis, TN, 38103, USA
| | - Kehkashan Parveen
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Arshi Parveen
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Michelle A Puchowicz
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Amandeep Bajwa
- Transplant Research Institute, College of Medicine, The University of Tennessee Health Science Center, 71 S Manassas St, Room 418H, Memphis, TN, 38103, USA.
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA.
- Neuroscience Institute, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
34
|
Perrone S, Carloni S, Dell'Orto VG, Filonzi L, Beretta V, Petrolini C, Lembo C, Buonocore G, Esposito S, Nonnis Marzano F. Hypoxic ischemic brain injury: animal models reveal new mechanisms of melatonin-mediated neuroprotection. Rev Neurosci 2024; 35:331-339. [PMID: 38153803 DOI: 10.1515/revneuro-2023-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/24/2023] [Indexed: 12/30/2023]
Abstract
Oxidative stress (OS) and inflammation play a key role in the development of hypoxic-ischemic (H-I) induced brain damage. Following H-I, rapid neuronal death occurs during the acute phase of inflammation, and activation of the oxidant-antioxidant system contributes to the brain damage by activated microglia. So far, in an animal model of perinatal H-I, it was showed that neuroprostanes are present in all brain damaged areas, including the cerebral cortex, hippocampus and striatum. Based on the interplay between inflammation and OS, it was demonstrated in the same model that inflammation reduced brain sirtuin-1 expression and affected the expression of specific miRNAs. Moreover, through proteomic approach, an increased expression of genes and proteins in cerebral cortex synaptosomes has been revealed after induction of neonatal H-I. Administration of melatonin in the experimental treatment of brain damage and neurodegenerative diseases has produced promising therapeutic results. Melatonin protects against OS, contributes to reduce the generation of pro-inflammatory factors and promotes tissue regeneration and repair. Starting from the above cited aspects, this educational review aims to discuss the inflammatory and OS main pathways in H-I brain injury, focusing on the role of melatonin as neuroprotectant and providing current and emerging evidence.
Collapse
Affiliation(s)
- Serafina Perrone
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Aurelio Saffi 2, 61029 Urbino, Italy
| | - Valentina Giovanna Dell'Orto
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Laura Filonzi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Virginia Beretta
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Petrolini
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Lembo
- Department of Neonatology, APHP, Necker-Enfants, Malades Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Via Banchi di Sotto 55, 53100 Siena, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Francesco Nonnis Marzano
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| |
Collapse
|
35
|
Zhang XY, Han PP, Zhao YN, Shen XY, Bi X. Crosstalk between autophagy and ferroptosis mediate injury in ischemic stroke by generating reactive oxygen species. Heliyon 2024; 10:e28959. [PMID: 38601542 PMCID: PMC11004216 DOI: 10.1016/j.heliyon.2024.e28959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Stroke represents a significant threat to global human health, characterized by high rates of morbidity, disability, and mortality. Predominantly, strokes are ischemic in nature. Ischemic stroke (IS) is influenced by various cell death pathways, notably autophagy and ferroptosis. Recent studies have increasingly highlighted the interplay between autophagy and ferroptosis, a process likely driven by the accumulation of reactive oxygen species (ROS). Post-IS, either the inhibition of autophagy or its excessive activation can escalate ROS levels. Concurrently, the interaction between ROS and lipids during ferroptosis further augments ROS accumulation. Elevated ROS levels can provoke endoplasmic reticulum stress-induced autophagy and, in conjunction with free iron (Fe2+), can trigger ferroptosis. Moreover, ROS contribute to protein and lipid oxidation, endothelial dysfunction, and an inflammatory response, all of which mediate secondary brain injury following IS. This review succinctly explores the mechanisms of ROS-mediated crosstalk between autophagy and ferroptosis and the detrimental impact of increased ROS on IS. It also offers novel perspectives for IS treatment strategies.
Collapse
Affiliation(s)
- Xing-Yu Zhang
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping-Ping Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yi-Ning Zhao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
36
|
Xu T, Chen T, Fang H, Shen X, Shen X, Tang Z, Zhao J. Human Umbilical Cord Mesenchymal Stem Cells Repair Endothelial Injury and Dysfunction by Regulating NLRP3 to Inhibit Endothelial Cell Pyroptosis in Kawasaki Disease. Inflammation 2024; 47:483-502. [PMID: 37948033 DOI: 10.1007/s10753-023-01921-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Vascular endothelial inflammation and endothelial dysfunction are the main causes of endothelial injury in Kawasaki disease (KD). Human umbilical cord-derived mesenchymal stem cells (Huc-MSCs) have multiple functions in immune regulation. This study examined whether Huc-MSCs inhibited endothelial inflammation and improved endothelial function in KD through constructing cell and in vivo animal KD vasculitis models. The pyroptosis factor NOD-like receptor protein 3 (NLRP3) was involved in the inflammatory process in the acute phase of KD. After tail vein injection of Huc-MSCs, inflammatory cell infiltration and the expression of pyroptosis-related proteins in the LCWE-induced KD mouse vasculitis model were significantly reduced. In vitro, NLRP3-dependent pyroptosis successfully induced human umbilical vein endothelial cell (HUVEC) damage. Huc-MSCs effectively increased the abilities of impaired HUVECs to proliferate, migrate, invade, and form vessel-like tubes, while inhibiting their apoptosis, suggesting that Huc-MSCs can reduce inflammation and improve vascular endothelial function by inhibiting the NLRP3-dependent pyroptosis pathway in KD, providing a possibility and novel target for KD endothelial injury and dysfunction.
Collapse
Affiliation(s)
- Ting Xu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Tao Chen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Hao Fang
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xiwei Shen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xianjuan Shen
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Jianmei Zhao
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China.
| |
Collapse
|
37
|
Igoe A, Twomey DM, Allen N, Carton S, Brady N, O'Keeffe F. A longitudinal analysis of factors associated with post traumatic growth after acquired brain injury. Neuropsychol Rehabil 2024; 34:430-452. [PMID: 37022203 DOI: 10.1080/09602011.2023.2195190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/20/2023] [Indexed: 04/07/2023]
Abstract
ABSTRACTPost-Traumatic Growth (PTG) is a form of positive psychological change that occurs for some individuals following traumatic experiences. High levels of PTG have been reported among survivors of acquired brain injury (ABI). Yet it remains unclear why some survivors of ABI develop PTG and others do not. The present study investigated early and late factors that are associated with long-term PTG in people with moderate to severe ABIs. Participants (n = 32, Mage = 50.59, SD = 12.28) completed self-report outcome measures at two time-points seven years apart (one-year and eight-years post-ABI). Outcome measures assessed emotional distress, coping, quality of life and ongoing symptoms of brain injury, as well as PTG at the later timepoint. Multiple regression analyses indicated that one-year post-ABI, fewer symptoms of depression, more symptoms of anxiety, and use of adaptive coping strategies accounted for a significant amount of variance in later PTG. At eight years post-ABI, fewer symptoms of depression, fewer ongoing symptoms of brain injury, better psychological quality of life and use of adaptive coping strategies explained a substantial amount of variance in PTG. For individuals with ABIs, PTG may be promoted by implementing long-term neuropsychological support which aims to facilitate use of adaptive coping strategies, supports psychological wellbeing and allows individuals to find meaning post-ABI.
Collapse
Affiliation(s)
- Anna Igoe
- University College Dublin, Dublin, Ireland
| | | | | | - Simone Carton
- National Rehabilitation Hospital, Dun Laoghaire, Ireland
| | | | - Fiadhnait O'Keeffe
- University College Dublin, Dublin, Ireland
- Trinity College Dublin, Dublin, Ireland
- National Rehabilitation Hospital, Dun Laoghaire, Ireland
| |
Collapse
|
38
|
Li Y, Li M, Feng S, Xu Q, Zhang X, Xiong X, Gu L. Ferroptosis and endoplasmic reticulum stress in ischemic stroke. Neural Regen Res 2024; 19:611-618. [PMID: 37721292 PMCID: PMC10581588 DOI: 10.4103/1673-5374.380870] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/30/2023] [Accepted: 06/14/2023] [Indexed: 09/19/2023] Open
Abstract
Ferroptosis is a form of non-apoptotic programmed cell death, and its mechanisms mainly involve the accumulation of lipid peroxides, imbalance in the amino acid antioxidant system, and disordered iron metabolism. The primary organelle responsible for coordinating external challenges and internal cell demands is the endoplasmic reticulum, and the progression of inflammatory diseases can trigger endoplasmic reticulum stress. Evidence has suggested that ferroptosis may share pathways or interact with endoplasmic reticulum stress in many diseases and plays a role in cell survival. Ferroptosis and endoplasmic reticulum stress may occur after ischemic stroke. However, there are few reports on the interactions of ferroptosis and endoplasmic reticulum stress with ischemic stroke. This review summarized the recent research on the relationships between ferroptosis and endoplasmic reticulum stress and ischemic stroke, aiming to provide a reference for developing treatments for ischemic stroke.
Collapse
Affiliation(s)
- Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Mingyang Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Shi Feng
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xu Zhang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
39
|
Boseley RE, Sylvain NJ, Peeling L, Kelly ME, Pushie MJ. A review of concepts and methods for FTIR imaging of biomarker changes in the post-stroke brain. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184287. [PMID: 38266967 DOI: 10.1016/j.bbamem.2024.184287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Stroke represents a core area of study in neurosciences and public health due to its global contribution toward mortality and disability. The intricate pathophysiology of stroke, including ischemic and hemorrhagic events, involves the interruption in oxygen and nutrient delivery to the brain. Disruption of these crucial processes in the central nervous system leads to metabolic dysregulation and cell death. Fourier transform infrared (FTIR) spectroscopy can simultaneously measure total protein and lipid content along with a number of key biomarkers within brain tissue that cannot be observed using conventional techniques. FTIR imaging provides the opportunity to visualize this information in tissue which has not been chemically treated prior to analysis, thus retaining the spatial distribution and in situ chemical information. Here we present a review of FTIR imaging methods for investigating the biomarker responses in the post-stroke brain.
Collapse
Affiliation(s)
- Rhiannon E Boseley
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Nicole J Sylvain
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Lissa Peeling
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Michael E Kelly
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - M Jake Pushie
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada.
| |
Collapse
|
40
|
Zhou Q, Ma J, Liu Q, Wu C, Yang Z, Yang T, Chen Q, Yue Y, Shang J. Traditional Chinese Medicine formula, Sanwujiao granule, attenuates ischemic stroke by promoting angiogenesis through early administration. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117418. [PMID: 37979814 DOI: 10.1016/j.jep.2023.117418] [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: 08/04/2023] [Revised: 10/24/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ischemic stroke (IS) is one of the most lethal diseases with the insufficient pharmacology therapeutic approach. Sanwujiao granule (SW) is widely used for IS in China with little known about its underlying mechanism. AIM OF THE STUDY To investigate the characteristics of therapeutic effects and potential mechanisms of SW against IS. MATERIALS AND METHODS The fingerprint of SW was applied by high-performance liquid chromatography-mass spectrometry (HPLC-MS). Three different drug treatment strategies, including prophylactic administration, early administration and delayed administration, were applied in rats' permanent middle cerebral occlusion (pMCAO) model. The Garcia neurological deficit test, adhesive removal test, rotarod test, TTC and TUNEL staining were performed to evaluate the pathological changes. The transcriptomic analysis was used to predict the potential mechanism of SW. The vascular deficiency model of Tg(kdrl:eGFP) zebrafish larvae and oxygen-glucose deprivation model on bEnd.3 cells were used to verify SW's pharmacological effect. qRT-PCR, immunofluorescent staining and Western Blot were applied to detect the expression of genes and proteins. The network pharmacology approach was applied to discover the potential bioactive compounds in SW that contribute to its pharmacological effect. RESULTS SW early and delayed administration attenuated cerebral infarction, neurological deficit and cell apoptosis. The transcriptomic analysis revealed that SW activated angiogenesis-associated biological processes specifically by early administration. CD31 immunofluorescent staining further confirmed the microvessel intensity in peri-infarct regions was significantly elevated after SW early treatment. Additionally, on the vascular deficiency model of zebrafish larvae, SW showed the angiogenesis effect. Next, the cell migration and tube formation were also observed in the bEnd.3 cells with the oxygen-glucose deprivation induced cell injury. It's worth noting that both mRNA and protein levels of angiogenesis factor, insulin-like growth factor 1, were significantly elevated in the pMCAO rats' brains treated with SW. The network pharmacology approach was applied and chasmanine, karacoline, talatisamine, etc. were probably the main active compounds of SW in IS treatment as they affected the angiogenesis-associated targets. CONCLUSIONS These results demonstrate that SW plays a critical role in anti-IS via promoting angiogenesis through early administration, indicating that SW is a candidate herbal complex for further investigation in treating IS in the clinical.
Collapse
Affiliation(s)
- Qinyang Zhou
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ji Ma
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Qiuyan Liu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Changyue Wu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ziwei Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Tingting Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Qimeng Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Yunyun Yue
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Jing Shang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 210009, China; NMPA Key Laboratory for Research and Evaluation of Cosmetics, National Institutes for Food and Drug Control, Beijing, 100050, China.
| |
Collapse
|
41
|
Yue C, Lian W, Duan M, Xia D, Cao X, Peng J. The predictive efficacy of programmed cell death in immunotherapy of melanoma: A comprehensive analysis of gene expression data for programmed cell death biomarker and therapeutic target discovery. ENVIRONMENTAL TOXICOLOGY 2024; 39:1858-1873. [PMID: 38140739 DOI: 10.1002/tox.24051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 12/24/2023]
Abstract
In this study, genes linked to prognosis in skin cutaneous melanoma (SKCM) involved in programmed cell death (PCD) were identified and confirmed and prognostic models based on these genes were constructed. Acquisition and analysis of clinical data and RNA sequencing information from The Cancer Genome Atlas-SKCM (TCGA-SKCM) and Sangerbox databases, gene expression data for 477 tumor samples and 2 normal samples were successfully gathered. The patients were separated into two clusters based on consensus clustering of PCD-related genes, with Cluster A having greater tumor purity, ESTIMATE score, immune score, and matrix score, and Cluster B having a significantly distinct pattern of immune cell infiltration. The use of gene set enrichment analysis and weighted correlation network analysis showed significant associations between certain genes and factors such as tumor mutation burden, age, stage, grade, and tumor subtype. Finally, based on the 12 genes selected by Least Absolute Shrinkage and Selection Operator regression analysis (STAT3, IRF2, SLC7A11, ZEB1, LIPT1, PML, GCH1, GYS1, ABCC1, XBP1, TFAP2C, NOX4), a prognostic model of PGD-related genes was constructed. The effectiveness of the model's prognostic value was confirmed through survival analysis, time-dependent receiver operating characteristic curve, single-factor Cox regression analysis, and nomogram. We also verified the relationship between the GCH1 and MKI67 expression by wet experiment. This model has high prediction accuracy in SKCM patients and can provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Chao Yue
- Department of Dermatologic Surgery, Hangzhou Third People's Hospital, Zhejiang, China
| | - Wenqin Lian
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Mengying Duan
- Department of Dermatologic Surgery, Hangzhou Third People's Hospital, Zhejiang, China
| | - Die Xia
- Department of medicine, China Medical University, Shenyang, China
| | - Xianbin Cao
- Department of Dermatologic Surgery, Hangzhou Third People's Hospital, Zhejiang, China
| | - Jianzhong Peng
- Department of Dermatologic Surgery, Hangzhou Third People's Hospital, Zhejiang, China
| |
Collapse
|
42
|
Jiang J, Qi T, Li L, Pan Y, Huang L, Zhu L, Zhang D, Ma X, Qin Y. MRPS9-Mediated Regulation of the PI3K/Akt/mTOR Pathway Inhibits Neuron Apoptosis and Protects Ischemic Stroke. J Mol Neurosci 2024; 74:23. [PMID: 38381220 DOI: 10.1007/s12031-024-02197-4] [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: 12/26/2023] [Accepted: 02/05/2024] [Indexed: 02/22/2024]
Abstract
Neuronal apoptosis is crucial in the pathophysiology of ischemic stroke (IS), albeit its underly24ing mechanism remaining elusive. Investigating the mechanism of neuronal apoptosis in the context of IS holds substantial clinical value for enhancing the prognosis of IS patients. Notably, the MRPS9 gene plays a pivotal role in regulating mitochondrial function and maintaining structural integrity. Utilizing bioinformatic tactics and the extant gene expression data related to IS, we conducted differential analysis and weighted correlation network analysis (WGCNA) to select important modules. Subsequent gene interaction analysis via the STRING website facilitated the identification of the key gene-mitochondrial ribosomal protein S9 (MRPS9)-that affects the progression of IS. Moreover, possible downstream signaling pathways, namely PI3K/Akt/mTOR, were elucidated via Kyoto Encyclopedia of Gene and Genomes (KEGG) and Gene Ontology (GO) pathway analysis. Experimental models were established utilizing oxygen-glucose deprivation/reoxygenation (OGD/R) in vitro and middle cerebral artery occlusion/reperfusion (MCAO/R) in mice. Changes in gene and protein expression, as well as cell proliferation and apoptosis, were monitored through qPCR, WB, CCK8, and flow cytometry. An OGD/R cell model was further employed to investigate the role of MRPS9 in IS post transfusion of MRPS9 overexpression plasmids into cells. Further studies were conducted by transfecting overexpressed cells with PI3K/Akt/mTOR signaling pathway inhibitor LY294002 to unveil the mechanism of MRPS9 in IS. Bioinformatic analysis revealed a significant underexpression of MRPS9 in ischemic stroke patients. Correspondingly, in vitro experiments with HN cells subjected to OGD/R treatment demonstrated a marked reduction in MRPS9 expression, accompanied by a decline in cell viability, and an increase cell apoptosis. Notably, the overexpression of MRPS9 mitigated the OGD/R-induced decrease in cell viability and augmentation of apoptosis. In animal models, MRPS9 expression was significantly lower in the MCAO/R group compared to the sham surgery group. Further, the KEGG pathway analysis associated MRPS9 expression with the PI3K/Akt/mTOR signaling pathway. In cells treated with the specific PI3K/Akt/mTOR inhibitor LY294002, phosphorylation levels of Akt and mTOR were decreased, cell viability decreased, and apoptosis increased compared to the MRPS9 overexpression group. These findings collectively indicate that MRPS9 overexpression inhibits PI3K/Akt/mTOR pathway activation, thereby protecting neurons from apoptosis and impeding IS progression. However, the PI3K/Akt/mTOR inhibitor LY294002 is capable of counteracting the protective effect of MRPS9 overexpression on neuronal apoptosis and IS. Our observations underscore the potential protective role of MRPS9 in modulating neuronal apoptosis and in attenuating the pathophysiological developments associated with IS. This is achieved through the regulation of the PI3K/Akt/mTOR pathway. These insights forge new perspectives and propose novel targets for the strategic diagnosis and treatment of IS.
Collapse
Affiliation(s)
- Jina Jiang
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Tingting Qi
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Li Li
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Yunzhi Pan
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Lijuan Huang
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Lijuan Zhu
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Dongyang Zhang
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Xiaoqing Ma
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China
| | - Yinghui Qin
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Tiefeng District, No. 3, Taishun Street, Qiqihar, China.
| |
Collapse
|
43
|
Teng C, Lv W, Chen Y, Liu L, Yin J, Li S, Min Z, Zhang Q, He W, Ma K, Li X, Cao X, Xin H. Enhanced the treatment of ischemic stroke through intranasal temperature-sensitive hydrogels of edaravone and borneol inclusion complex. Int J Pharm 2024; 651:123748. [PMID: 38154533 DOI: 10.1016/j.ijpharm.2023.123748] [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: 10/24/2023] [Revised: 11/29/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Since ischemic stroke occurs by a combination of multiple mechanisms, therapies that modulate multiple mechanisms are required for its treatment. The combination of edaravone (EDA) and borneol can significantly ameliorate the symptoms of neurological deficits in cerebral ischemia-reperfusion model in rats. In this study, the solubility of borneol and edaravone was improved by hydroxypropyl-β-cyclodextrin and PEG400. Furthermore, a nasal temperature-sensitive hydrogel containing both edaravone and borneol inclusion complex (EDA-BP TSGS) was developed to overcome the obstacles of ischemic stroke treatment including the obstruction of the blood-brain barrier (BBB) and the unavailability and untimely of intravenous injection. The effectiveness of the thermosensitive hydrogel was investigated in transient middle cerebral artery occlusion/reperfusion model rats (MCAO/R). The results showed that EDA-BP TSGS could significantly alleviate the symptoms of neurological deficits and decrease the cerebral infarct area and the degree of brain damage. In summary, nasal EDA-BP TSGS is a secure and effective brain-targeting formulation that may provide a viable option for the clinical prophylaxis and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chuanhui Teng
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wei Lv
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Wuxi 214400, China
| | - Yuqin Chen
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Linfeng Liu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jiaqing Yin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Shengnan Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhiyi Min
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qi Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Weichong He
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Kunfang Ma
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xinrui Li
- Department of Pharmacy, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China.
| | - Xiang Cao
- Department of Pharmacy, Kangda College of Nanjing Medical University, Lianyungang 222000, China.
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
44
|
Peng C, Ai Q, Zhao F, Li H, Sun Y, Tang K, Yang Y, Chen N, Liu F. Quercetin attenuates cerebral ischemic injury by inhibiting ferroptosis via Nrf2/HO-1 signaling pathway. Eur J Pharmacol 2024; 963:176264. [PMID: 38123006 DOI: 10.1016/j.ejphar.2023.176264] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
AIMS Ischemic stroke is a severe cerebrovascular disease in which neuronal death continually occurs through multiple forms, including apoptosis, autophagy, pyroptosis and ferroptosis. Quercetin (QRC), as a natural flavonoid compound, has been reported to have pharmacological effects on ischemic injury accompanied by unclear anti-ferroptotic mechanisms. This study is designed to investigate the therapeutic effects of QRC against ferroptosis in ischemic stroke. MATERIALS AND METHODS In vivo, the model of MCAO rats were used to assess the protective effect of QRC on cerebral ischemic. Additionally, we constructed oxidative stressed and ferroptotic cell models to explore the effects and mechanisms of QRC on ferroptosis. The related proteins were analysed by western blotting, immunohistochemical and immunofluorescence techniques. RESULTS The experiments demonstrated that QRC improves neurological deficits, infarct volume, and pathological features in MCAO rats, also increased the viability of HT-22 cells exposed to H2O2 and erastin. These results, including MDA, SOD, GSH, ROS levels and iron accumulation, indicated that QRC suppresses the generation of lipid peroxides and may involve in the regulatory of ferroptosis. Both in vitro and in vivo, QRC was found to inhibit ferroptosis by up-regulating GPX4 and FTH1, as well as down-regulating ACSL4. Furthermore, we observed that QRC enhances the nuclear translocation of Nrf2 and activates the downstream antioxidative proteins. Importantly, the effect of QRC on ferroptosis can be reversed by the Nrf2 inhibitor ML385. CONCLUSIONS This study provides evidence that QRC has a neuroprotective effect by inhibiting ferroptosis, demonstrating the therapeutic potential for cerebral ischemic stroke.
Collapse
Affiliation(s)
- Caiwang Peng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, 410208, China
| | - Qidi Ai
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China
| | - Fengyan Zhao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, 410208, China
| | - Hengli Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, 410208, China
| | - Yang Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China
| | - Keyan Tang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, 410208, China
| | - Yantao Yang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China.
| | - Naihong Chen
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Fang Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China; Center for Standardization and Functional Engineering of Traditional Chinese Medicine in Hunan Province, Changsha, 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, 410208, China.
| |
Collapse
|
45
|
Turovsky EA, Baryshev AS, Plotnikov EY. Selenium Nanoparticles in Protecting the Brain from Stroke: Possible Signaling and Metabolic Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:160. [PMID: 38251125 PMCID: PMC10818530 DOI: 10.3390/nano14020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
Strokes rank as the second most common cause of mortality and disability in the human population across the world. Currently, available methods of treating or preventing strokes have significant limitations, primarily the need to use high doses of drugs due to the presence of the blood-brain barrier. In the last decade, increasing attention has been paid to the capabilities of nanotechnology. However, the vast majority of research in this area is focused on the mechanisms of anticancer and antiviral effects of nanoparticles. In our opinion, not enough attention is paid to the neuroprotective mechanisms of nanomaterials. In this review, we attempted to summarize the key molecular mechanisms of brain cell damage during ischemia. We discussed the current literature regarding the use of various nanomaterials for the treatment of strokes. In this review, we examined the features of all known nanomaterials, the possibility of which are currently being studied for the treatment of strokes. In this regard, the positive and negative properties of nanomaterials for the treatment of strokes have been identified. Particular attention in the review was paid to nanoselenium since selenium is a vital microelement and is part of very important and little-studied proteins, e.g., selenoproteins and selenium-containing proteins. An analysis of modern studies of the cytoprotective effects of nanoselenium made it possible to establish the mechanisms of acute and chronic protective effects of selenium nanoparticles. In this review, we aimed to combine all the available information regarding the neuroprotective properties and mechanisms of action of nanoparticles in neurodegenerative processes, especially in cerebral ischemia.
Collapse
Affiliation(s)
- Egor A. Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia
| | - Alexey S. Baryshev
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991 Moscow, Russia;
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
46
|
Motawi TK, Sadik NAH, Shaker OG, Ghaleb MMH, Elbaz EM. Expression, Functional Polymorphism, and Diagnostic Values of MIAT rs2331291 and H19 rs217727 Long Non-Coding RNAs in Cerebral Ischemic Stroke Egyptian Patients. Int J Mol Sci 2024; 25:842. [PMID: 38255915 PMCID: PMC10815378 DOI: 10.3390/ijms25020842] [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: 11/18/2023] [Revised: 12/24/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Cerebral ischemic stroke (CIS) is a severe cerebral vascular event. This research aimed to evaluate the role of single-nucleotide polymorphisms (SNPs) of the lncRNAs MIAT rs2331291 and H19 rs217727 and epigenetic methylation in the expression patterns of serum lncRNA H19 in CIS Egyptian patients. It included 80 CIS cases and 40 healthy subjects. Serum MIAT expression levels decreased, whereas serum H19 expression levels increased among CIS compared to controls. For MIAT rs2331291, there were significant differences in the genotypic and allelic frequencies between the CIS and healthy subjects at p = 0.02 and p = 0.0001, respectively. Our findings illustrated a significantly increased MIAT T/T genotype frequency in hypertensive CIS compared to non-hypertensive CIS at p = 0.004. However, H19 rs217727 gene frequency C/C was not significantly higher in non-hypertensive CIS than in hypertensive CIS. The methylation of the H19 gene promoter was significantly higher in CIS patients compared to healthy subjects. The level of MIAT was positively correlated with serum H19 in CIS. Receiver operating characteristics (ROC) analysis revealed that serum MIAT and H19 have a high diagnostic potential for distinguishing CIS subjects from healthy ones. In conclusion, the MIAT-rs2331291 polymorphism might serve as a novel potential indicator of CIS.
Collapse
Affiliation(s)
- Tarek K. Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | | | - Olfat G. Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | | | - Eman M. Elbaz
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
47
|
Ya J, Pellumbaj J, Hashmat A, Bayraktutan U. The Role of Stem Cells as Therapeutics for Ischaemic Stroke. Cells 2024; 13:112. [PMID: 38247804 PMCID: PMC10814781 DOI: 10.3390/cells13020112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Stroke remains one of the leading causes of death and disability worldwide. Current reperfusion treatments for ischaemic stroke are limited due to their narrow therapeutic window in rescuing ischaemic penumbra. Stem cell therapy offers a promising alternative. As a regenerative medicine, stem cells offer a wider range of treatment strategies, including long-term intervention for chronic patients, through the reparation and replacement of injured cells via mechanisms of differentiation and proliferation. The purpose of this review is to evaluate the therapeutic role of stem cells for ischaemic stroke. This paper discusses the pathology during acute, subacute, and chronic phases of cerebral ischaemic injury, highlights the mechanisms involved in mesenchymal, endothelial, haematopoietic, and neural stem cell-mediated cerebrovascular regeneration, and evaluates the pre-clinical and clinical data concerning the safety and efficacy of stem cell-based treatments. The treatment of stroke patients with different types of stem cells appears to be safe and efficacious even at relatively higher concentrations irrespective of the route and timing of administration. The priming or pre-conditioning of cells prior to administration appears to help augment their therapeutic impact. However, larger patient cohorts and later-phase trials are required to consolidate these findings.
Collapse
Affiliation(s)
| | | | | | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neurosciences, Queens Medical Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
48
|
Zhu T, Liu H, Gao S, Jiang N, Chen S, Xie W. Effect of salidroside on neuroprotection and psychiatric sequelae during the COVID-19 pandemic: A review. Biomed Pharmacother 2024; 170:115999. [PMID: 38091637 DOI: 10.1016/j.biopha.2023.115999] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has affected the mental health of individuals worldwide, and the risk of psychiatric sequelae and consequent mental disorders has increased among the general population, health care workers and patients with COVID-19. Achieving effective and widespread prevention of pandemic-related psychiatric sequelae to protect the mental health of the global population is a serious challenge. Salidroside, as a natural agent, has substantial pharmacological activity and health effects, exerts obvious neuroprotective effects, and may be effective in preventing and treating psychiatric sequelae and mental disorders resulting from stress stemming from the COVID-19 pandemic. Herein, we systematically summarise, analyse and discuss the therapeutic effects of salidroside in the prevention and treatment of psychiatric sequelae as well as its roles in preventing the progression of mental disorders, and fully clarify the potential of salidroside as a widely applicable agent for preventing mental disorders caused by stress; the mechanisms underlying the potential protective effects of salidroside are involved in the regulation of the oxidative stress, neuroinflammation, neural regeneration and cell apoptosis in the brain, the network homeostasis of neurotransmission, HPA axis and cholinergic system, and the improvement of synaptic plasticity. Notably, this review innovatively proposes that salidroside is a potential agent for treating stress-induced health issues during the COVID-19 pandemic and provides scientific evidence and a theoretical basis for the use of natural products to combat the current mental health crisis.
Collapse
Affiliation(s)
- Ting Zhu
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Hui Liu
- Guizhou Provincial Key Laboratory of Pharmaceutics & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550004, Guizhou, China; Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Shiman Gao
- Department of Clinical Pharmacy, Women and Children's Hospital, Qingdao University, Qingdao 266034, China
| | - Ning Jiang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
| | - Shuai Chen
- School of Public Health, Wuhan University, Donghu Road No. 115, Wuchang District, Wuhan 430071, China.
| | - Weijie Xie
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai 200122, China.
| |
Collapse
|
49
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [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: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Goel D, Shangari S, Mittal M, Bhat A. Endogenous defense mechanism-based neuroprotection in large-vessel acute ischemic stroke: A hope for future. Brain Circ 2024; 10:51-59. [PMID: 38655439 PMCID: PMC11034449 DOI: 10.4103/bc.bc_56_23] [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: 06/13/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Stroke is a leading cause of morbidity and mortality worldwide and a leading cause of disability. None of the neuroprotective agents have been approved internationally except edaravone in Japanese guidelines in acute ischemic stroke. We here discuss that there are two types of endogenous defense mechanisms (EDMs) after acute stroke for neuromodulation and neuroregeneration, and if both can be activated simultaneously, then we can have better recovery in stroke. AIMS AND OBJECTIVES We aimed to study the effect of combination of neuroprotection therapies acting on the two wings of EDM in acute large-vessel middle cerebral artery (LMCA) ischemic stroke. METHODS Sixty patients of LMCA stroke were enrolled and randomized within 72 h into two groups of 30 patients each. The control group received standard medical care without any neuroprotective agents while the intervention group received standard medical care combined with oral citicoline with vinpocetine for 3 months with initial 1 week intravenous and edaravone and cerebrolysin injection, started within 72 h of onset of stroke. Patients were assessed on the basis of the National Institutes of Health Stroke Scale, Fugl-Meyer Assessment Score, Glasgow Coma Scale, and Mini-Mental Status Examination at admission, discharge, and after 90 days. RESULTS The intervention group showed significant and early improvements in motor as well as cognitive recovery. CONCLUSION Combination therapy for neuroprotection which is acting on two pathways of EDM can be useful in functional recovery after acute ischemic stroke.
Collapse
Affiliation(s)
- Deepak Goel
- Department of Neurology, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| | - Sushant Shangari
- Department of Neurology, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| | - Manish Mittal
- Department of Neurology, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| | - Ashwani Bhat
- Department of Neurology, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| |
Collapse
|