1
|
Zhao A, Zhang G, Wei H, Yan X, Gan J, Jiang X. Heat shock proteins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic implications. Exp Neurol 2025; 390:115284. [PMID: 40318821 DOI: 10.1016/j.expneurol.2025.115284] [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: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) remains a significant challenge in ischemic stroke treatment. Heat shock proteins (HSPs), a cadre of molecular chaperones, have emerged as pivotal regulators in this pathological cascade. This review synthesizes the latest research on HSPs in CIRI from 2013 to 2024 focusing on their multifaceted roles and therapeutic potential. We explore the diverse cellular functions of HSPs, including regulation of oxidative stress, apoptosis, necroptosis, ferroptosis, autophagy, neuroinflammation, and blood-brain barrier integrity. Key HSPs, such as HSP90, HSP70, HSP32, HSP60, HSP47, and small HSPs, are investigated for their specific mechanisms of action in CIRI. Potential therapeutic strategies targeting HSPs, including HSP inhibitors, traditional Chinese medicine components, and gene therapy, are discussed. This review provides a comprehensive understanding of HSPs in CIRI and offers insights into the development of innovative neuroprotective treatments.
Collapse
Affiliation(s)
- Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
Shu H, Liao Q, Chen Z, Liang M, Zhang S, Liu J, Wu Y, Hu P, Luo M, Zhu W, Zhu X, Yang L, Yan T. Flavonoids serve as a promising therapeutic agent for ischemic stroke. Brain Res 2025; 1853:149528. [PMID: 39999903 DOI: 10.1016/j.brainres.2025.149528] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Ischemic stroke (IS) continues to be a major public health concern and is characterized by significantly high mortality and disabling rates. Inhibiting nerve cells death and enhancing the repair of ischemic tissue are important treatment concepts for IS. Currently, the mainstream treatment strategies mainly focus on short-term care, which underscores the urgent need for novel therapeutic strategies for long-term care. Emerging data reveal that flavonoids have surfaced as promising candidates for IS patients' long-term care. Flavonoids can alleviate neuroinflammation and anti-apoptosis due to their characteristic pharmacological mechanisms. Clinical evidence suggests that long-term flavonoids intake improves IS patients' long-term outcomes. Though the effect of flavonoids in IS treatment has been explored for decades, the neuroprotective pharmacodynamics have not been well established. Thereby, the aim of current review is to summarize the pathways involved in neuroprotective effect of flavonoids. This review will also advance the potential of flavonoids as a viable clinical candidate for the treatment of IS.
Collapse
Affiliation(s)
- Hongxin Shu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiuye Liao
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihao Chen
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Mingyu Liang
- School of life sciences, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si Zhang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junzhe Liu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yanze Wu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ping Hu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming Luo
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenping Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Li Yang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tengfeng Yan
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
3
|
Barati A, Moghimi S, Taghavi Zanjani K, Rohani M, Sohrabi Hesar M, Arfaie A, Ghezelche Khamsiyan M, Mahmoudi J, Sadigh-Eteghad S. Acute Administration of Edaravone Improves Cognitive Impairment in a Mouse Model of mPFC Ischemia: Crosstalk Between Necroptosis, Neuroinflammation, and Antioxidant Defense. Mol Neurobiol 2025; 62:4420-4434. [PMID: 39448519 DOI: 10.1007/s12035-024-04541-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
Edaravone (Eda), a well-known free radical scavenger, has been reported as a possible therapeutic agent for ischemic stroke patients' recovery. This study aimed to investigate the effects of time-dependent treatment with Eda on medial prefrontal cortex (mPFC) ischemia. Mice were randomly allocated into six groups: control, sham, normal saline, Eda-I, Eda-II, and Eda-III. After induction of a photothrombotic ischemia in the mPFC region, Eda-I, Eda-II, and Eda-III groups received 3 mg/kg Eda intraperitoneally at the times of 0, 2, and 6 h post-surgery. After 1 day of recovery, the mice underwent behavioral tests (open field, novel object recognition, and T-maze). Next, necroptosis, NOD-like receptor protein 3 (NLRP3), and nuclear factor erythroid 2-related factor 2 (Nrf2) pathway-related protein levels were measured in the lesioned area using western blot analysis. For double confirmation, IL-1β and IL-18 were also assessed by immunofluorescence in the area. Further, histological evaluations were performed to measure tissue damage. The results showed that mPFC ischemia impaired recognition and spatial working memory without affecting locomotor activity, while immediate Eda administration improved cognitive impairments. Furthermore, acute Eda treatment reduced RIP1, RIP3, and MLKL levels, inhibited NLRP3 inflammasome proteins (NLRP3, ASC, and Cas1), decreased IL-1β and IL-18, upregulated Nrf2 and its targets (NQO-1 and HO-1), and diminished tissue damage. Our results highlighted the effects of acute administration of Eda post-stroke on improving cognitive impairments by suppressing necroptosis and NLRP3 inflammasome pathways and activating the Nrf2 antioxidant defense mechanism.
Collapse
Affiliation(s)
- Alireza Barati
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadegh Moghimi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kiana Taghavi Zanjani
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mojde Rohani
- Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Mehri Sohrabi Hesar
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Arian Arfaie
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Shadman J, Haghi-Aminjan H, Alipour MR, Panahpour H. The Neuroprotective Mechanisms of Kaempferol in Experimental Ischemic Stroke: A Preclinical Systematic Review. Mol Neurobiol 2025:10.1007/s12035-025-04848-y. [PMID: 40120044 DOI: 10.1007/s12035-025-04848-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Ischemic stroke represents a critical global health challenge, resulting in significant mortality and disability worldwide, yet there are limited effective treatment options currently available. While the intricate molecular pathways underlying the onset and progression of ischemic stroke are multifaceted, relying on a single therapeutic approach is unlikely to yield effective treatment for this complex disease. Therefore, it is crucial to explore efficient strategies that employ multifaceted targeting and address the multifarious pathological processes to overcome the challenges associated with ischemic brain injury. In recent times, natural plant-derived compounds have garnered significant interest as promising neuroprotective agents for the management of neurological conditions, including ischemic stroke. This study investigates the possible neuroprotective properties of kaempferol, a naturally occurring flavonoid compound, in mitigating the detrimental consequences of cerebral ischemic events. The findings from the reviewed preclinical studies suggest that kaempferol exhibits significant neuroprotective potential as a multifaceted therapeutic agent for ischemic stroke. Its efficacy stems from a combination of antioxidant, anti-inflammatory, and anti-apoptotic properties, which collectively mitigate ischemic stroke-induced brain injury. While these results are promising, clinical studies are essential to validate kaempferol's therapeutic viability for ischemic stroke patients.
Collapse
Affiliation(s)
- Javad Shadman
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | | | - Hamdollah Panahpour
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
5
|
Long S, Wang Y. Association of TAB2 gene polymorphism with endometrial cancer susceptibility and clinical analysis. Turk J Obstet Gynecol 2025; 22:1-12. [PMID: 40062608 PMCID: PMC11894771 DOI: 10.4274/tjod.galenos.2025.24983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 03/14/2025] Open
Abstract
Objective Transforming growth factor-β-activated kinase 1 binding protein 2 (TAB2) plays a vital role in inflammatory pathways. It has also been considered a potential target for the enhancement of the the antiestrogen effects. Previous evidence has indicated that TAB2 gene variants are associated with several diseases, whereas their potential correlation with endometrial cancer (EC) is unclear. This study aims to initially explore the association between TAB2 gene polymorphisms (rs237028 /AG, rs521845 T/G, and rs652921 T/C) and EC. Materials and Methods Polymerase chain reaction-restriction fragment length polymorphism was applied to determine the genotype composition and the allele frequencies of TAB2 gene variant polymorphisms in 270 EC patients and 294 healthy controls. Results The G allele of rs521845 was related to the increase of EC risk [p=0.08, odds ratio (OR): 0.72, 95% confidence interval (CI): 0.56-0.91]. Moreover, EC risk was associated with rs521845 in different genetic models (p=0.017, OR: 0.63, 95% CI: 0.44-0.91 in the codominant model; p=0.0051, OR: 0.61, 95% CI: 0.43-0.87 in the dominant model). For rs237028, the percentage of AG genotype in patients with highly differentiated tumours (G1) was significantly higher than that in moderately, poorly differentiated patients (G2/G3) (p=0.031, OR: 0.77, 95% CI: 0.45-1.30). Conclusion Our results showed that the rs521845 polymorphism of TAB2, was associated with EC risk, suggesting that TAB2 may play a crucial role in EC prognosis.
Collapse
Affiliation(s)
- Siyu Long
- Sichuan University, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Center for Translational Medicine, Laboratory of Molecular Translational Medicine, Sichuan, China
- Sichuan University West China Second University Hospital, Clinic of Andrology/Sichuan Human Sperm Bank, Chengdu, China
| | - Yanyun Wang
- Sichuan University, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Center for Translational Medicine, Laboratory of Molecular Translational Medicine, Sichuan, China
| |
Collapse
|
6
|
Azami P, Mohammadzadeh S, Seirafi S, Razeghian-Jahromi I. A review of cutting-edge biomarkers for diagnosing coronary artery disease. Medicine (Baltimore) 2025; 104:e41377. [PMID: 39854741 PMCID: PMC11771658 DOI: 10.1097/md.0000000000041377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Chronic coronary artery disease (CAD) remains a significant global healthcare burden. Current risk assessment methods have notable limitations in early detection and risk stratification. Hence, there is an urgent need for innovative biomarkers that facilitate the premature CAD diagnosis, ultimately leading to reduction in associated morbidity and mortality rates. This review comprehensively examines recent advances in emerging biomarkers for CAD detection. Our analysis delves into various aspects of these biomarkers such as their mechanisms of action, roles in the pathophysiology of the disease, and different measurement techniques employed in clinical practice. Comparative assessment of biomarker performance between CAD patients and control groups was also presented relying on their sensitivity, specificity, and area under the curve at specific cutoff points. In this regard, prominent biomarkers including Tenascin-C, IL-37, PTX3, transthyretin, soluble interleukin-6 receptor α, and miR-15a are identified as having high diagnostic potential for chronic CAD that indeed showcase promising performance metrics. These findings underscore the role of novel biomarkers in enhancing CAD risk stratification and improving patient outcomes through early intervention. However, the pursuit of an ideal and inclusive biomarker continues due to the multifaceted nature of CAD. Future randomized controlled trials are essential to bridge the gap between research findings and clinical practice in order to augment the practical application of these biomarkers in routine healthcare settings.
Collapse
Affiliation(s)
- Pouria Azami
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Soroush Seirafi
- Department of Cardiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
7
|
Lee J, Geum D, Park DH, Kim JH. Molecular Targeting of Ischemic Stroke: The Promise of Naïve and Engineered Extracellular Vesicles. Pharmaceutics 2024; 16:1492. [PMID: 39771472 PMCID: PMC11678501 DOI: 10.3390/pharmaceutics16121492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and long-term disability worldwide, with limited therapeutic options available. Despite the success of early interventions, such as tissue-type plasminogen activator administration and mechanical thrombectomy, many patients continue to experience persistent neurological deficits. The pathophysiology of IS is multifaceted, encompassing excitotoxicity, oxidative and nitrosative stress, inflammation, and blood-brain barrier disruption, all of which contribute to neural cell death, further complicating the treatment of IS. Recently, extracellular vesicles (EVs) secreted naturally by various cell types have emerged as promising therapeutic agents because of their ability to facilitate selective cell-to-cell communication, neuroprotection, and tissue regeneration. Furthermore, engineered EVs, designed to enhance targeted delivery and therapeutic cargo, hold the potential to improve their therapeutic benefits by mitigating neuronal damage and promoting neurogenesis and angiogenesis. This review summarizes the characteristics of EVs, the molecular mechanisms underlying IS pathophysiology, and the emerging role of EVs in IS treatment at the molecular level. This review also explores the recent advancements in EV engineering, including the incorporation of specific proteins, RNAs, or pharmacological agents into EVs to enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Jihun Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea;
| | - Dongho Geum
- Department of Medical Science, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Dong-Hyuk Park
- Department of Neurosurgery, Anam Hospital, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea;
| |
Collapse
|
8
|
Shilenok I, Kobzeva K, Soldatov V, Deykin A, Bushueva O. C11orf58 (Hero20) Gene Polymorphism: Contribution to Ischemic Stroke Risk and Interactions with Other Heat-Resistant Obscure Chaperones. Biomedicines 2024; 12:2603. [PMID: 39595169 PMCID: PMC11592265 DOI: 10.3390/biomedicines12112603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Recently identified Hero proteins, which possess chaperone-like functions, are promising candidates for research into atherosclerosis-related diseases, including ischemic stroke (IS). Methods: 2204 Russian subjects (917 IS patients and 1287 controls) were genotyped for fifteen common SNPs in Hero20 gene C11orf58 using probe-based PCR and the MassArray-4 system. Results: Six C11orf58 SNPs were significantly associated with an increased risk of IS in the overall group (OG) and significantly modified by smoking (SMK) and low fruit/vegetable intake (LFVI): rs10766342 (effect allele (EA) A; P(OG = 0.02; SMK = 0.009; LFVI = 0.04)), rs11024032 (EA T; P(OG = 0.01; SMK = 0.01; LFVI = 0.036)), rs11826990 (EA G; P(OG = 0.007; SMK = 0.004; LFVI = 0.03)), rs3203295 (EA C; P(OG = 0.016; SMK = 0.01; LFVI = 0.04)), rs10832676 (EA G; P(OG = 0.006; SMK = 0.002; LFVI = 0.01)), rs4757429 (EA T; P(OG = 0.02; SMK = 0.04; LFVI = 0.04)). The top ten intergenic interactions of Hero genes (two-, three-, and four-locus models) involved exclusively polymorphic loci of C11orf58 and C19orf53 and were characterized by synergic and additive (independent) effects between SNPs. Conclusions: Thus, C11orf58 gene polymorphism represents a major risk factor for IS. Bioinformatic analysis showed the involvement of C11orf58 SNPs in molecular mechanisms of IS mediated by their role in the regulation of redox homeostasis, inflammation, vascular remodeling, apoptosis, vasculogenesis, neurogenesis, lipid metabolism, proteostasis, hypoxia, cell signaling, and stress response. In terms of intergenic interactions, C11orf58 interacts most closely with C19orf53.
Collapse
Affiliation(s)
- Irina Shilenok
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Division of Neurology, Kursk Emergency Hospital, 305035 Kursk, Russia
| | - Ksenia Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Vladislav Soldatov
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Alexey Deykin
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| |
Collapse
|
9
|
Muench NA, Schmitt HM, Schlamp CL, Su AJA, Washington K, Nickells RW. Preservation of Murine Whole Eyes With Supplemented UW Cold Storage Solution: Anatomical Considerations. Transl Vis Sci Technol 2024; 13:24. [PMID: 39560629 DOI: 10.1167/tvst.13.11.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024] Open
Abstract
Purpose Retinal ganglion cell (RGC) apoptosis and axon regeneration are the principal obstacles challenging the development of successful whole eye transplantation (WET). The purpose of this study was to create a neuroprotective cocktail that targets early events in the RGC intrinsic apoptotic program to stabilize RGCs in a potential donor eye. Methods University of Wisconsin (UW) solution was augmented with supplements known to protect RGCs. Supplements targeted tyrosine kinase signaling, histone deacetylase activity, K+ ion efflux, macroglial stasis, and provided energy support. Modified UW (mUW) solutions with individual supplements were injected into the vitreous of enucleated mouse eyes, which were then stored in cold UW solution for 24 hours. Histopathology, immunostaining of individual retinal cell types, and analysis of cell-specific messenger RNAs (mRNAs) were used to identify supplements that were combined to create optimal mUW solution. Results UW and mUW solutions reduced ocular edema and focal ischemia in globes stored in cold storage. Two major issues were noted after cold storage, including retinal detachment and reduction in glial fibrillary acidic protein staining in astrocytes. A combination of supplements resolved both these issues and performed better than the individual supplements alone. Cold storage resulted in a reduction in cell-specific mRNAs, even though it preserved the corresponding protein products. Conclusions Eyes treated with optimal mUW solution exhibited preservation of retinal and cellular architecture, but did display a decrease in mRNA levels, suggesting that cold storage induced cellular stasis. Translational Relevance Application of optimal mUW solution lowers an important barrier to the development of a successful whole eye transplantation procedure.
Collapse
Affiliation(s)
- Nicole A Muench
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Heather M Schmitt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Perfuse Therapeutics Inc., Durham, NC, USA
| | - Cassandra L Schlamp
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - An-Jey A Su
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kia Washington
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- https://orcid.org/0000-0002-2998-5494
| |
Collapse
|
10
|
Yan W, Wang C, Zhao Y, Jiang Y, Sun M. Involvement of Calpain in Neurovascular Unit Damage through Up-regulating PARP-NF-κB Signaling during Experimental Ischemic Stroke. Mol Neurobiol 2024; 61:8104-8122. [PMID: 38472651 DOI: 10.1007/s12035-024-04092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/03/2024] [Indexed: 03/14/2024]
Abstract
Calpain and PARP-NF-κB signaling are reported to participate in the ischemic brain injury. In this study, it was investigated whether calpain was contributed to the neurovascular unit (NVU) damage through up-regulating PARP-NF-κB signaling during experimental ischemic stroke. Male Sprague-Dawley rats were suffered from 90 min of middle cerebral artery occlusion, followed by reperfusion. The NVU damage was evaluated by the permeability of blood-brain barrier (BBB), the degradation of proteins in extracellular matrix and tight junctions, and ultrastructural changes. The inflammatory response was determined by the expression of inflammatory genes driven by PARP-NF-κB signaling and the activities of myeloperoxidase (MPO). Treatment with MDL 28,170, a calpain inhibitor, improved neurological functions, reduced TUNEL staining index, lessened brain swelling, and decreased infarct volume in ischemic rats. Moreover, it reduced the BBB permeability, enhanced the levels of laminin, collagen IV and occludin, and attenuated the ultrastructural damage of NVU in penumbra and core after induction of ischemia. Meanwhile, it enhanced the levels of cytosolic IκBα, lessened the levels of nuclear PARP and NF-κB p65, reduced the levels of ICAM-1, TNF-α, IL-1β, MMP-9, and MMP-2,and suppressed the activities of MPO in penumbra and core. These data showed that calpain inhibition suppressed PARP-NF-κB signaling-mediated inflammatory response, reduced NVU damage, and protected brain against ischemic stroke, suggesting the involvement of calpain in the NVU damage through up-regulating PARP-NF-κB signaling during brain ischemia.
Collapse
Affiliation(s)
- Wenhao Yan
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Chunyang Wang
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yumei Zhao
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yingying Jiang
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Ming Sun
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
11
|
Li R, Lou Q, Ji T, Li Y, Yang H, Ma Z, Zhu Y, Qian C, Yang W, Wang Y, Luo S. Mechanism of Astragalus mongholicus Bunge ameliorating cerebral ischemia-reperfusion injury: Based on network pharmacology analysis and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118157. [PMID: 38588987 DOI: 10.1016/j.jep.2024.118157] [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: 01/16/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus mongholicus Bunge (AMB) is a herb with wide application in traditional Chinese medicine, exerting a wealth of pharmacological effects. AMB has been proven to have an evident therapeutic effect on ischemic cerebrovascular diseases, including cerebral ischemia-reperfusion injury (CIRI). However, the specific mechanism underlying AMB in CIRI remains unclear. AIM OF THE STUDY This study aimed to investigate the potential role of AMB in CIRI through a comprehensive approach of network pharmacology and in vivo experimental research. METHODS The intersection genes of drugs and diseases were obtained through analysis of the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and Gene Expression Omnibus (GEO) database. The protein-protein interaction (PPI) network was created through the string website. Meanwhile, the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was carried out using R studio, and thereafter the key genes were screened. Then, the molecular docking prediction was made between the main active ingredients and target genes, and hub genes with high binding energy were obtained. In addition, molecular dynamic (MD) simulation was used to validate the result of molecular docking. Based on the results of network pharmacology, we used animal experiments to verify the predicted hub genes. First, the rat middle cerebral artery occlusion and reperfusion (MACO/R) model was established and the effective dose of AMB in CIRI was determined by behavioral detection and 2,3,5-Triphenyltetrazolium chloride (TTC) staining. Then the target proteins corresponding to the hub genes were measured by Western blot. Moreover, the level of neuronal death was measured using hematoxylin and eosin (HE) and Nissl staining. RESULTS Based on the analysis of the TCMSP database and GEO database, a total of 62 intersection target genes of diseases and drugs were obtained. The KEGG enrichment analysis showed that the therapeutic effect of AMB on CIRI might be realized through the advanced glycation endproduct-the receptor of advanced glycation endproduct (AGE-RAGE) signaling pathway in diabetic complications, nuclear factor kappa-B (NF-κB) signaling pathway and other pathways. Molecular docking results showed that the active ingredients of AMB had good binding potential with hub genes that included Prkcb, Ikbkb, Gsk3b, Fos and Rela. Animal experiments showed that AWE (60 g/kg) could alleviate CIRI by regulating the phosphorylation of PKCβ, IKKβ, GSK3β, c-Fos and NF-κB p65 proteins. CONCLUSION AMB exerts multi-target and multi-pathway effects against CIRI, and the underlying mechanism may be related to anti-apoptosis, anti-inflammation, anti-oxidative stress and inhibiting calcium overload.
Collapse
Affiliation(s)
- Rui Li
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, 230061, PR China
| | - Qi Lou
- Department of Pharmacology, School of Basic Medicine, Anhui Medical University, Hefei, 230031, PR China
| | - Tingting Ji
- Department of Pharmacy, Anhui Medical College, Hefei, 230601, PR China
| | - Yincan Li
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, PR China
| | - Haoran Yang
- Department of Pharmacy, Anhui Medical College, Hefei, 230601, PR China
| | - Zheng Ma
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, 230061, PR China
| | - Yu Zhu
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, 230061, PR China
| | - Can Qian
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, 230061, PR China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China.
| | - Yijun Wang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, PR China.
| | - Shengyong Luo
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, 230061, PR China; Department of Pharmacology, School of Basic Medicine, Anhui Medical University, Hefei, 230031, PR China.
| |
Collapse
|
12
|
Bonetti M, Giugno L, Borsani E, Bonomini F. Potential Neuroprotective Effect of Melatonin in the Hippocampus of Male BTBR Mice. Nutrients 2024; 16:1652. [PMID: 38892585 PMCID: PMC11174678 DOI: 10.3390/nu16111652] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder identified by impairments in common social interactions and repetitive behaviors. In ASD patients, substantial morphological alterations have been observed in the hippocampus, which represents an important region for the development of social skills. Melatonin, commonly found in many foods and plants, is also produced by the pineal gland. This indolamine, known to regulate the circadian rhythm, shows antioxidant and anti-inflammatory properties. We therefore hypothesized that melatonin may reduce oxidative stress and inflammation in the hippocampus of ASD patients. We explored our hypothesis using the BTBR mouse, a well-regarded murine transgenic model for ASD. Immediately after weaning, male BTBR and C57BL/6 mice underwent an 8-week treatment with melatonin or vehicle. Later, through immunohistochemistry and the immunoblotting analysis of the hippocampus, we evaluated the overall expression and cellular localization of Nrf2 and SOD1, two enzymes involved in the oxidative stress response. Similarly, we evaluated NLRP3 and NFkB, two mediators of inflammation, and GAD67, an enzyme responsible for the synthesis of GABA. Ultimately, we addressed melatonin's potential to regulate iron metabolism through a DAB-enhanced Perls reaction assay. Results showed melatonin's potential for modulating the analyzed markers in BTBR mice, suggesting a potential neuroprotective effect in ASD patients.
Collapse
Affiliation(s)
- Matteo Bonetti
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.B.); (L.G.); (E.B.)
| | - Lorena Giugno
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.B.); (L.G.); (E.B.)
| | - Elisa Borsani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.B.); (L.G.); (E.B.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Francesca Bonomini
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.B.); (L.G.); (E.B.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
13
|
Xu M, Qian LH, Wang JX, He ZY, Ling XY, Wang WH, Wang JW, Hu Y, Gong MJ. Rutaecarpine Alleviates Early Brain Injury-Induced Inflammatory Response Following Subarachnoid Hemorrhage via SIRT6/NF-[Formula: see text]B Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:799-819. [PMID: 38752843 DOI: 10.1142/s0192415x24500320] [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: 05/31/2024]
Abstract
Subarachnoid hemorrhage (SAH), a specific subtype of cerebrovascular accident, is characterized by the extravasation of blood into the interstice between the brain and its enveloping delicate tissues. This pathophysiological phenomenon can precipitate an early brain injury (EBI), which is characterized by inflammation and neuronal death. Rutaecarpine (Rut), a flavonoid compound discovered in various plants, has been shown to have protective effects against SAH-induced cerebral insult in rodent models. In our study, we used a rodent SAH model to evaluate the effect of Rut on EBI and investigated the effect of Rut on the inflammatory response and its regulation of SIRT6 expression in vitro. We found that Rut exerts a protective effect on EBI in SAH rats, which is partly due to its ability to inhibit the inflammatory response. Notably, Rut up-regulated Sirtuin 6 (SIRT6) expression, leading to an increase in H3K9 deacetylation and inhibition of nuclear factor-kappa B (NF-[Formula: see text]B) transcriptional activation, thereby mediating the inflammatory response. In addition, further data showed that SIRT6 was proven to mediate the regulation of Rut on the microglial inflammatory response. These findings highlight the importance of SIRT6 in the regulation of inflammation and suggest a potential mechanism for the protective effect of Rut on EBI. In summary, Rut may have the potential to prevent and treat SAH-induced brain injury by interacting with SIRT6. Our findings may provide a new therapeutic strategy for the treatment of SAH-induced EBI.
Collapse
Affiliation(s)
- Min Xu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Li-Hui Qian
- School of Medicine, Nanjing University of Chinese Medicine 210023, Nanjing, P. R. China
| | - Jun-Xiang Wang
- Department of Neurosurgery, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University 215500, Jiangsu Province, P. R. China
| | - Zi-Yang He
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Xiao-Yang Ling
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Wen-Hua Wang
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Jin-Wen Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine 210023, Nanjing, P. R. China
| | - Yue Hu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine 210023, Nanjing, P. R. China
- Shen Chun-Ti Nation-Famous Experts Studio for Traditional Chinese Medicine Inheritance, Changzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou 213003, Jiangsu, P. R. China
- Department of Neurology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210001, P. R. China
| | - Ming-Jie Gong
- Department of Neurosurgery, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University 215500, Jiangsu Province, P. R. China
| |
Collapse
|
14
|
Zhang L, Zhou X, Zhao J, Wang X. Research hotspots and frontiers of preconditioning in cerebral ischemia: A bibliometric analysis. Heliyon 2024; 10:e24757. [PMID: 38317957 PMCID: PMC10839892 DOI: 10.1016/j.heliyon.2024.e24757] [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: 07/11/2023] [Revised: 12/13/2023] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
Background Preconditioning is a promising strategy against ischemic brain injury, and numerous studies in vitro and in vivo have demonstrated its neuroprotective effects. However, at present there is no bibliometric analysis of preconditioning in cerebral ischemia. Therefore, a comprehensive overview of the current status, hot spots, and emerging trends in this research field is necessary. Materials and methods Studies on preconditioning in cerebral ischemia from January 1999-December 2022 were retrieved from the Web of Science Core Collection (WOSCC) database. CiteSpace was used for data mining and visual analysis. Results A total of 1738 papers on preconditioning in cerebral ischemia were included in the study. The annual publications showed an upwards and then downwards trend but currently remain high in terms of annual publications. The US was the leading country, followed by China, the most active country in recent years. Capital Medical University published the largest number of articles. Perez-Pinzon, Miguel A contributed the most publications, while KITAGAWA K was the most cited author. The focus of the study covered three areas: (1) relevant diseases and experimental models, (2) types of preconditioning and stimuli, and (3) mechanisms of ischemic tolerance. Remote ischemic preconditioning, preconditioning of mesenchymal stem cells (MSCs), and inflammation are the frontiers of research in this field. Conclusion Our study provides a visual and scientific overview of research on preconditioning in cerebral ischemia, providing valuable information and new directions for researchers.
Collapse
Affiliation(s)
- Long Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Traditional Chinese Medicine, Zibo TCM-Integrated Hospital, Zibo ,255026, China
| | - Xue Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jing Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xingchen Wang
- Division of Neurology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China
| |
Collapse
|
15
|
Chen Y, Zhang C, Zhao L, Chen R, Zhang P, Li J, Zhang X, Zhang X. Eriocalyxin B alleviated ischemic cerebral injury by limiting microglia-mediated excessive neuroinflammation in mice. Exp Anim 2024; 73:124-135. [PMID: 37839867 PMCID: PMC10877152 DOI: 10.1538/expanim.23-0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023] Open
Abstract
Excessive neuroinflammation mediated by microglia has a detrimental effect on the progression of ischemic stroke. Eriocalyxin B (EriB) was found with a neuroprotective effect in mice with Parkinson's disease via the suppression of microglial overactivation. This study aimed to investigate the roles of EriB in permanent middle cerebral artery occlusion (pMCAO) mice. The pMCAO was induced in the internal carotid artery of the mice by the intraluminal filament method, and EriB (10 mg/kg) was administered immediately after surgery by intraperitoneal injection. The behavior score, 2,3,5-triphenyltetrazole chloride staining, Nissl staining, TUNEL, immunohistochemistry, immunofluorescence, PCR, ELISA, and immunoblotting revealed that EriB administration reduced brain infarct and neuron death and ameliorated neuroinflammation and microglia overactivation in pMCAO mice, manifested by alterations of TUNEL-positive cell numbers, ionized calcium binding adaptor molecule 1 (Iba-1)-positive cell numbers, and expression of tumor necrosis factor-α, interleukin 6, IL-1β, inducible nitric oxide synthase, and arginase 1. In addition, EriB suppressed ischemia-induced activation of nuclear factor kappa B (NF-κB) signaling in the brain penumbra, suggesting the involvement of NF-κB in EriB function. In conclusion, EriB exerted anti-inflammatory effects in ischemia stroke by regulating the NF-κB signaling pathway, and this may provide insights into the neuroprotective effect of EriB in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yanqiang Chen
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- Department of Neurology, Hebei Chest Hospital, 372 Shengli North Street, Shijiazhuang, 050000, Hebei, P.R. China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 309 Zhonghua North Street, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
| | - Liming Zhao
- Department of Neurology, Hebei Chest Hospital, 372 Shengli North Street, Shijiazhuang, 050000, Hebei, P.R. China
| | - Rong Chen
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 309 Zhonghua North Street, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
| | - Peipei Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 309 Zhonghua North Street, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
| | - Junxia Li
- Department of Neurology, Hebei Chest Hospital, 372 Shengli North Street, Shijiazhuang, 050000, Hebei, P.R. China
| | - Xueping Zhang
- Department of Neurology, Hebei Chest Hospital, 372 Shengli North Street, Shijiazhuang, 050000, Hebei, P.R. China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 309 Zhonghua North Street, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 215 Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei, P.R. China
| |
Collapse
|
16
|
Xu G, Dong F, Su L, Tan ZX, Lei M, Li L, Wen D, Zhang F. The role and therapeutic potential of nuclear factor κB (NF-κB) in ischemic stroke. Biomed Pharmacother 2024; 171:116140. [PMID: 38211425 DOI: 10.1016/j.biopha.2024.116140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Stroke is a prevalent cerebrovascular condition with a global impact, causing significant rates of illness and death. Despite extensive research, the available treatment options for stroke remain restricted. Hence, it is crucial to gain a deeper understanding of the molecular mechanisms associated with the onset and advancement of stroke in order to establish a theoretical foundation for novel preventive and therapeutic approaches. NF-κB, also known as nuclear factor κB, is a transcription factor responsible for controlling the expression of numerous genes and plays a crucial role in diverse physiological processes. NF-κB is triggered and regulates neuroinflammation and other processes after stroke, promoting the generation of cytokine storms and contributing to the advancement of ischemic stroke (IS). Therefore, NF-κB could potentially play a vital role in stroke by regulating diverse pathophysiological processes. This review provides an overview of the functions of NF-κB in stroke and its governing mechanisms. In addition, our attention is directed towards various potential therapies that aim to inhibit the NF-κB signaling pathway in order to offer valuable insights for the advancement of innovative treatment approaches for stroke.
Collapse
Affiliation(s)
- Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lei Su
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Mingcheng Lei
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lina Li
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, PR China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, PR China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China.
| |
Collapse
|
17
|
Mirzahosseini G, Ismael S, Salman M, Kumar S, Ishrat T. Genetic and Pharmacological Modulation of P75 Neurotrophin Receptor Attenuate Brain Damage After Ischemic Stroke in Mice. Mol Neurobiol 2024; 61:276-293. [PMID: 37606717 DOI: 10.1007/s12035-023-03550-1] [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: 01/19/2023] [Accepted: 07/29/2023] [Indexed: 08/23/2023]
Abstract
The precursor nerve growth factor (ProNGF) and its receptor p75 neurotrophin receptor (p75NTR) are upregulated in several brain diseases, including ischemic stroke. The activation of p75NTR is associated with neuronal apoptosis and inflammation. Thus, we hypothesized that p75NTR modulation attenuates brain damage and improves functional outcomes after ischemic stroke. Two sets of experiments were performed. (1) Adult wild-type (WT) C57BL/6 J mice were subjected to intraluminal suture-middle cerebral artery occlusion (MCAO) to induce cerebral ischemia. Pharmacological inhibitor of p75NTR, LM11A-31 (50 mg/kg), or normal saline was administered intraperitoneally (IP) 1 h post-MCAO, and animals survived for 24 h. (2) Adult p75NTR heterozygous knockout (p75NTR+/-) and WT were subjected to photothrombotic (pMCAO) to induce ischemic stroke, and the animals survived for 72 h. The sensory-motor function of animals was measured using Catwalk XT. The brain samples were collected to assess infarction volume, edema, hemorrhagic transformation, neuroinflammation, and signaling pathway at 24 and 72 h after the stroke. The findings described that pharmacological inhibition and genetic knocking down of p75NTR reduce infarction size, edema, and hemorrhagic transformation following ischemic stroke. Additionally, p75NTR modulation significantly decreased several anti-apoptosis markers and improved sensory motor function compared to the WT mice following ischemic stroke. Our observations exhibit that the involvement of p75NTR in ischemic stroke and modulation of p75NTR could improve the outcome of ischemic stroke by increasing cell survival and enhancing motor performance. LM11A-31 has the potential to be a promising therapeutic agent for ischemic stroke. However, more evidence is needed to illuminate the efficacy of LM11A-31 in ischemic stroke.
Collapse
Affiliation(s)
- Golnoush Mirzahosseini
- 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, TN, 38163, Memphis, USA
| | - Saifudeen Ismael
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, LA, 70112, New Orleans, USA
| | - 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
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, TN, 38163, Memphis, 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, TN, 38163, Memphis, USA.
- Neuroscience Institute, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
18
|
Sethiya NK, Ghiloria N, Srivastav A, Bisht D, Chaudhary SK, Walia V, Alam MS. Therapeutic Potential of Myricetin in the Treatment of Neurological, Neuropsychiatric, and Neurodegenerative Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:865-882. [PMID: 37461364 DOI: 10.2174/1871527322666230718105358] [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: 08/11/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 06/12/2024]
Abstract
Myricetin (MC), 3,5,7,3',4',5'-hexahydroxyflavone, chemically belongs to a flavonoid category known to confer antioxidant, antimicrobial, antidiabetic, and neuroprotective effects. MC is known to suppress the generation of Reactive Oxygen Species (ROS), lipid peroxidation (MDA), and inflammatory markers. It has been reported to improve insulin function in the human brain and periphery. Besides this, it modulates several neurochemicals including glutamate, GABA, serotonin, etc. MC has been shown to reduce the expression of the enzyme Mono Amine Oxidase (MAO), which is responsible for the metabolism of monoamines. MC treatment reduces levels of plasma corticosterone and restores hippocampal BDNF (full form) protein in stressed animals. Further, MC has shown its protective effect against amyloid-beta, MPTP, rotenone, 6-OHDA, etc. suggesting its potential role against neurodegenerative disorders. The aim of the present review is to highlight the therapeutic potential of MC in the treatment of several neurological, neuropsychiatric, and neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Neha Ghiloria
- Dr. Baba Saheb Ambedkar Hospital, Rohini, New Delhi 110085, India
| | | | - Dheeraj Bisht
- Department of Pharmaceutical Sciences, Sir J.C. Bose Technical Campus, Bhimtal, Kumaun University, Nainital, Uttarakhand 263002, India
| | | | - Vaibhav Walia
- Department of Pharmacology, SGT College of Pharmacy, SGT University, Gurugram, Haryana 122505, India
| | - Md Sabir Alam
- Department of Pharmaceutics, SGT College of Pharmacy, SGT University, Gurugram, Haryana 122505, India
| |
Collapse
|
19
|
Tournier BB, Sorce S, Marteyn A, Ghidoni R, Benussi L, Binetti G, Herrmann FR, Krause K, Zekry D. CCR5 deficiency: Decreased neuronal resilience to oxidative stress and increased risk of vascular dementia. Alzheimers Dement 2024; 20:124-135. [PMID: 37489764 PMCID: PMC10917026 DOI: 10.1002/alz.13392] [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: 12/24/2022] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
INTRODUCTION As the chemokine receptor5 (CCR5) may play a role in ischemia, we studied the links between CCR5 deficiency, the sensitivity of neurons to oxidative stress, and the development of dementia. METHODS Logistic regression models with CCR5/apolipoprotein E (ApoE) polymorphisms were applied on a sample of 205 cognitively normal individuals and 189 dementia patients from Geneva. The impact of oxidative stress on Ccr5 expression and cell death was assessed in mice neurons. RESULTS CCR5-Δ32 allele synergized with ApoEε4 as risk factor for dementia and specifically for dementia with a vascular component. We confirmed these results in an independent cohort from Italy (157 cognitively normal and 620 dementia). Carriers of the ApoEε4/CCR5-Δ32 genotype aged ≥80 years have an 11-fold greater risk of vascular-and-mixed dementia. Oxidative stress-induced cell death in Ccr5-/- mice neurons. DISCUSSION We propose the vulnerability of CCR5-deficient neurons in response to oxidative stress as possible mechanisms contributing to dementia.
Collapse
Affiliation(s)
- Benjamin B. Tournier
- Department of PsychiatryGeneva University Hospitals and University of GenevaGenevaSwitzerland
| | - Silvia Sorce
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Antoine Marteyn
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
- Division of GeriatricsDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
- Division of Internal Medicine for the AgedDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| | - Roberta Ghidoni
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Luisa Benussi
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - François R Herrmann
- Division of GeriatricsDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| | - Karl‐Heinz Krause
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Dina Zekry
- Division of Internal Medicine for the AgedDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| |
Collapse
|
20
|
Zou H, Chen X, Lu J, Zhou W, Zou X, Wu H, Li Z, Zhou X. Neurotropin alleviates cognitive impairment by inhibiting TLR4/MyD88/NF-κB inflammation signaling pathway in mice with vascular dementia. Neurochem Int 2023; 171:105625. [PMID: 37774797 DOI: 10.1016/j.neuint.2023.105625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Vascular dementia (VD) is the second most common cause of dementia after Alzheimer's disease. Neuroinflammation contributes to pathogenesis of VD. Neurotropin (NTP) is an analgesic that has been shown to suppress inflammation and neural repair. But its effects on VD are still unclear. Therefore, this study aimed to investigate the therapeutic effects and potential mechanisms of NTP in the VD model mice established by bilateral common carotid artery stenosis method. In VD mice, we found that NTP treatment increased cerebral blood flow by Laser speckle imaging, reduced neuron loss by Nissl, HE and immunochemistry staining, attenuated white matter damage by magnetic resonance imaging and ultrastructural damage by transmission electron microscope, improved cognitive functions by new object recognition test and three-chamber test, Y maze test and Morris water maze test, inhibited significantly glial activation by immunofluorescence methods, reduced the expression of TLR4, down-regulated expression of MyD88 and phosphorylation of NF-κB P65, decreased the levels of pro-inflammatory cytokines IL-1β, IL-6 and TNFα. Further, we showed that administration of a TLR4 inhibitor TAK242 had a similar effect to NTP, while the TLR4 agonist CRX-527 attenuated the effect of NTP in the VD mice. Collectively, our study suggested that NTP alleviates cognitive impairment by inhibiting TLR4/MyD88/NF-κB inflammation signaling pathway in the VD mice. Thus, NTP may be a promising therapeutic approach and a potential TLR4 inhibitor for VD.
Collapse
Affiliation(s)
- Huihui Zou
- Special Medical Service Center, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China
| | - Xinrun Chen
- Department of Neurology, General Hospital of Southern Theater Command, Chinese People's Liberation Army, Guangzhou, China
| | - Jiancong Lu
- Special Medical Service Center, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China
| | - Wanfei Zhou
- Special Medical Service Center, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China
| | - Xiaopei Zou
- Special Medical Service Center, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China
| | - Heyong Wu
- Special Medical Service Center, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China
| | - Zhou Li
- Department of Intensive Care Unit, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Xianju Zhou
- Special Medical Service Center, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| |
Collapse
|
21
|
Shen Z, Huang D, Jia N, Zhao S, Pei C, Wang Y, Wu Y, Wang X, Shi S, Wang F, He Y, Wang Z. Protective effects of Eleutheroside E against high-altitude pulmonary edema by inhibiting NLRP3 inflammasome-mediated pyroptosis. Biomed Pharmacother 2023; 167:115607. [PMID: 37776644 DOI: 10.1016/j.biopha.2023.115607] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
Eleutheroside E (EE) is a primary active component of Acanthopanax senticosus, which has been reported to inhibit the expression of inflammatory genes, but the underlying mechanisms remain elusive. High-altitude pulmonary edema (HAPE) is a severe complication of high-altitude exposure occurring after ascent above 2500 m. However, effective and safe preventative measures for HAPE still need to be improved. This study aimed to elucidate the preventative potential and underlying mechanism of EE in HAPE. Rat models of HAPE were established through hypobaric hypoxia. Mechanistically, hypobaric hypoxia aggravates oxidative stress and upregulates (pro)-inflammatory cytokines, activating NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis, eventually leading to HAPE. EE suppressed NLRP3 inflammasome-mediated pyroptosis by inhibiting the nuclear translocation of nuclear factor kappa-Β (NF-κB), thereby protecting the lung from HAPE. However, nigericin (Nig), an NLRP3 activator, partially abolished the protective effects of EE. These findings suggest EE is a promising agent for preventing HAPE induced by NLRP3 inflammasome-mediated pyroptosis.
Collapse
Affiliation(s)
- Zherui Shen
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Demei Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Nan Jia
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Sijing Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Caixia Pei
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yilan Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yongcan Wu
- Chongqing Medical University, Chongqing 400016, China
| | - Xiaomin Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shihua Shi
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Fei Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| | - Yacong He
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; State Key Laboratory of Southwestern Chinese Medicine Resources School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Zhenxing Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
22
|
Eom SH, Hong GL, Kang HB, Lee NS, Kim DK, Jeong YG, Kim CS, Yoo YC, Lee BH, Jung JY, Kim DS, Han SY. Neuroprotective Effects of Water Extract from Brown Algae Petalonia binghamiae in an Experimental Model of Focal Cerebral Ischemia In Vitro and In Vivo. Curr Issues Mol Biol 2023; 45:8427-8443. [PMID: 37886974 PMCID: PMC10605114 DOI: 10.3390/cimb45100531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Focal cerebral ischemia (fCI) can result in brain injury and sensorimotor deficits. Brown algae are currently garnering scientific attention as potential therapeutic candidates for fCI. This study investigated the therapeutic effects of the hot water extract of Petalonia binghamiae (wPB), a brown alga, in in vitro and in vivo models of fCI. The neuroprotective efficacy of wPB was evaluated in an in vitro excitotoxicity model established using HT-22 cells challenged with glutamate. Afterward, C57/BL6 mice were administered wPB for 7 days (10 or 100 mg/kg, intragastric) and subjected to middle cerebral artery occlusion and reperfusion (MCAO/R) operation, which was used as an in vivo fCI model. wPB co-incubation significantly inhibited cell death, oxidative stress, and apoptosis, as well as stimulated the expression of heme oxygenase-1 (HO-1), an antioxidant enzyme, and the nuclear translocation of its upstream regulator, nuclear factor erythroid 2-related factor 2 (Nrf2) in HT-22 cells challenged with glutamate-induced excitotoxicity. Pretreatment with either dose of wPB significantly attenuated infarction volume, neuronal death, and sensorimotor deficits in an in vivo fCI model. Furthermore, the attenuation of oxidative stress and apoptosis in the ischemic lesion accompanied the wPB-associated protection. This study suggests that wPB can counteract fCI via an antioxidative effect, upregulating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Sun Ho Eom
- Healinnols Inc., Daejeon 34054, Republic of Korea; (S.H.E.); (H.B.K.)
| | - Geum-Lan Hong
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea (N.-S.L.); (D.K.K.); (Y.G.J.)
| | - Hyun Bae Kang
- Healinnols Inc., Daejeon 34054, Republic of Korea; (S.H.E.); (H.B.K.)
| | - Nam-Seob Lee
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea (N.-S.L.); (D.K.K.); (Y.G.J.)
| | - Do Kyung Kim
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea (N.-S.L.); (D.K.K.); (Y.G.J.)
| | - Young Gil Jeong
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea (N.-S.L.); (D.K.K.); (Y.G.J.)
| | - Chun-Sung Kim
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Republic of Korea;
| | - Yung Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea;
| | - Bong Ho Lee
- Department of Chemical Technology, Hanbat National University, Daejeon 34158, Republic of Korea;
| | - Ju-Young Jung
- Department of Histology & Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Dong-Sub Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Gwangju 61473, Republic of Korea;
| | - Seung Yun Han
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea (N.-S.L.); (D.K.K.); (Y.G.J.)
| |
Collapse
|
23
|
Wei L, Li X, Wei Q, Chen L, Xu L, Zhou P. Oxidative Stress-mediated Sprouty-related Protein with an EVH1 Domain 1 Down-regulation Contributes to Resisting Oxidative Injury in Microglia. Neuroscience 2023; 526:13-20. [PMID: 37343716 DOI: 10.1016/j.neuroscience.2023.06.012] [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: 02/22/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023]
Abstract
Microglia play an ambiguous role in injury or repair after ischemia-reperfusion, and the induced oxidative stress serves as an important signal, mediates direct toxicity to nerve cells, and eventually simulates complex physiological processes such as activation of microglia to repair the damaged area. Herein, we show that sprouty-related protein with an EVH1 domain 1 (SPRED1) may act as a regulatory node in this phenomenon. The ischemic brain of an ischemia-reperfusion rat model constructed by middle cerebral artery occlusion (MCAO) showed an increase in oxidative stress and downregulation of SPRED1 expression. Hydrogen peroxide (H2O2)-simulated oxidative damage exerted a fluctuating regulatory effect on SPRED1 level in BV2 microglia, which is highly consistent with its regulatory effect on nuclear factor kappa B (NF-κB) transcription factor p65. Interestingly, SPRED1 overexpressed in BV2 cells did not exert any regulatory effect on p38 mitogen-activated protein kinase (MAPK), NF-κB p65, and pro-inflammatory cytokines. However, treatment of BV2 cells overexpressing SPRED1 with H2O2 led to significant changes in the above phenomena as well as their viability and apoptosis. In the absence of H2O2 induction, SPRED overexpression alone did not mediate such an effect. These findings indicate that SPRED1 tends to maintain intracellular homeostasis of signals, but the oxidative stress derived from ischemia-reperfusion can easily degrade SPRED1 and consequently re-activate these restricted signals and alter the behavior of microglia. Thus, our study reveals a novel role of SPRED1 in microglia in response to cerebral ischemia-induced oxidative stress.
Collapse
Affiliation(s)
- Li Wei
- Department of Blood Transfusion, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xin Li
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qianfeng Wei
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lin Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Li Xu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Peng Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
24
|
Howell JA, Gaouette N, Lopez M, Burke SP, Perkins E, Bidwell GL. Elastin-like polypeptide delivery of anti-inflammatory peptides to the brain following ischemic stroke. FASEB J 2023; 37:e23077. [PMID: 37402128 PMCID: PMC10349587 DOI: 10.1096/fj.202300309rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Inflammatory processes are activated following ischemic stroke that lead to increased tissue damage for weeks following the ischemic insult, but there are no approved therapies that target this inflammation-induced secondary injury. Here, we report that SynB1-ELP-p50i, a novel protein inhibitor of the nuclear factor kappa B (NF-κB) inflammatory cascade bound to the drug carrier elastin-like polypeptide (ELP), decreases NF-κB induced inflammatory cytokine production in cultured macrophages, crosses the plasma membrane and accumulates in the cytoplasm of both neurons and microglia in vitro, and accumulates at the infarct site where the blood-brain barrier (BBB) is compromised following middle cerebral artery occlusion (MCAO) in rats. Additionally, SynB1-ELP-p50i treatment reduces infarct volume by 11.86% compared to saline-treated controls 24 h following MCAO. Longitudinally, SynB1-ELP-p50i treatment improves survival for 14 days following stroke with no effects of toxicity or peripheral organ dysfunction. These results show high potential for ELP-delivered biologics for therapy of ischemic stroke and other central nervous system disorders and further support targeting inflammation in ischemic stroke.
Collapse
Affiliation(s)
- John Aaron Howell
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Nicholas Gaouette
- School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Mariper Lopez
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Stephen P. Burke
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
25
|
Li B, Zhang B, Li Z, Li S, Li J, Wang A, Hou J, Xu J, Zhang R. Ginkgolide C attenuates cerebral ischemia/reperfusion-induced inflammatory impairments by suppressing CD40/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116537. [PMID: 37094696 DOI: 10.1016/j.jep.2023.116537] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/06/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginkgo biloba L. (Ginkgoaceae), a traditional Chinese medicine, has been applied for thousands of years for the treatment of cardio-cerebral vascular diseases in China. It is written in Compendium of Materia Medica that Ginkgo has the property of "dispersing poison", which is now referred to as anti-inflammatory and antioxidant. Ginkgolides are important active ingredients in Ginkgo biloba leaves and ginkgolide injection has been frequently applied in clinical practice for the treatment of ischemic stroke. However, few studies have explored the effect and mechanism of ginkgolide C (GC) with anti-inflammatory activity in cerebral ischemia/reperfusion injury (CI/RI). AIM OF THE STUDY The present study aimed to demonstrate whether GC was capable of attenuating CI/RI. Furthermore, the anti-inflammatory effect of GC in CI/RI was explored around the CD40/NF-κB pathway. MATERIALS AND METHODS In vivo, middle cerebral artery occlusion/reperfusion (MCAO/R) model was established in rats. The neuroprotective effect of GC was assessed by neurological scores, cerebral infarct rate, microvessel ultrastructure, blood-brain barrier (BBB) integrity, brain edema, neutrophil infiltration, and levels of TNF-α, IL-1β, IL-6, ICAM-1, VCAM-1, and iNOS. In vitro, rat brain microvessel endothelial cells (rBMECs) were preincubated in GC before hypoxia/reoxygenation (H/R) culture. The cell viability, levels of CD40, ICAM-1, MMP-9, TNF-α, IL-1β, and IL-6, and activation of NF-κB pathway were examined. In addition, the anti-inflammatory effect of GC was also investigated by silencing CD40 gene in rBMECs. RESULTS GC attenuated CI/RI as demonstrated by decreasing neurological scores, reducing cerebral infarct rate, improving microvessel ultrastructural features, ameliorating BBB disruption, attenuating brain edema, inhibiting MPO activity, and downregulating levels of TNF-α, IL-1β, IL-6, ICAM-1, VCAM-1, and iNOS. Coherently, in rBMECs exposed to H/R GC enhanced cell viability and downregulated levels of ICAM-1, MMP-9, TNF-α, IL-1β, and IL-6. Furthermore, GC suppressed CD40 overexpression and hindered translocation of NF-κB p65 from the cytosol to the nucleus, phosphorylation of IκB-α, and activation of IKK-β in H/R rBMECs. However, GC failed to protect rBMECs from H/R-induced inflammatory impairments and suppress activation of NF-κB pathway when CD40 gene was silenced. CONCLUSIONS GC attenuates cerebral ischemia/reperfusion-induced inflammatory impairments by suppressing CD40/NF-κB pathway, which may provide an available therapeutic drug for CI/RI.
Collapse
Affiliation(s)
- Bin Li
- Graduate Department, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China; Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Baoke Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhenyu Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shasha Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jun Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Aiwu Wang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jinling Hou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jiping Xu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Rui Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
26
|
Li X, Gao Y, Han X, Tang S, Li N, Liu X, Ni X. Maresin1 ameliorates postoperative cognitive dysfunction in aged rats by potentially regulating the NF-κB pathway to inhibit astrocyte activation. Exp Gerontol 2023; 176:112168. [PMID: 37055002 DOI: 10.1016/j.exger.2023.112168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is one of the most serious postoperative complications in the elderly population. Perioperative central neuroinflammation is considered to be an important pathological mechanism of POCD, with the activation of astrocytes playing a key role in central neuroinflammation. Maresin1 (MaR1) is a specific pro-resolving mediator synthesized by macrophages in the resolution stage of inflammation, and provides unique anti-inflammatory and pro-resolution effects by limiting excessive neuroinflammation and promoting postoperative recovery. However, the question remains whether MaR1 can have a positive effect on POCD. The objective of this study was to investigate the protective effect of MaR1 on POCD cognitive function in aged rats after splenectomy. Morris water maze test and IntelliCage test showed that splenectomy could cause transient cognitive dysfunction in aged rats; however, the cognitive impairment of rats was significantly mitigated when MaR1 pretreatment was administered. MaR1 significantly alleviated the fluorescence intensity and protein expression of glial fibrillary acidic protein and central nervous system specific protein in the cornu ammonis 1 region of the hippocampus. Simultaneously, the morphology of astrocytes was also severely altered. Further experiments showed that MaR1 inhibited the mRNA and protein expression of several key proinflammatory cytokines-interleukin-1β, interleukin-6, and tumor necrosis factor-α in the hippocampus of aged rats following splenectomy. The molecular mechanism underlying this process was explored by evaluating expression of components of the nuclear factor κB (NF-κB) signaling pathway. MaR1 substantially inhibited the mRNA and protein expression of NF-κB p65 and κB inhibitor kinase β. Collectively, these results suggest that MaR1 ameliorated splenectomy-induced transient cognitive impairment in elderly rats, and this neuroprotective mechanism may occur through regulating the NF-κB pathway to inhibit astrocyte activation.
Collapse
Affiliation(s)
- Xiuhua Li
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yubo Gao
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xu Han
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Shaling Tang
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Na Li
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xing Liu
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xinli Ni
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
27
|
Howell JA, Gaouette N, Lopez M, Burke SP, Perkins E, Bidwell GL. Elastin-like polypeptide delivery of anti-inflammatory peptides to the brain following ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532834. [PMID: 36993686 PMCID: PMC10055169 DOI: 10.1101/2023.03.15.532834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Inflammatory processes are activated following ischemic strokes and lead to increased tissue damage for weeks following the ischemic insult, but there are no approved therapies that target this inflammation-induced secondary injury. Here, we report that SynB1-ELP-p50i, a novel protein inhibitor of the nuclear factor kappa B (NF-κB) inflammatory cascade bound to drug carrier elastin-like polypeptide (ELP), is able to enter both neurons and microglia, cross the blood-brain barrier, localize exclusively in the ischemic core and penumbra in Wistar-Kyoto and spontaneously hypertensive rats (SHRs), and reduce infarct volume in male SHRs. Additionally, in male SHRs, SynB1-ELP-p50i treatment improves survival for 14 days following stroke with no effects of toxicity or peripheral organ dysfunction. These results show high potential for ELP-delivered biologics for therapy of ischemic stroke and other central nervous system disorders and further support targeting inflammation in ischemic stroke.
Collapse
Affiliation(s)
- John Aaron Howell
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Nicholas Gaouette
- School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Mariper Lopez
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Stephen P. Burke
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
28
|
Low vitamin D status is associated with inflammatory response in older patients with cerebral small vessel disease. J Neuroimmunol 2023; 377:578057. [PMID: 36921477 DOI: 10.1016/j.jneuroim.2023.578057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/18/2023] [Accepted: 02/25/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVES This study aimed to determine the association of the NF-κB inflammatory signaling pathway with vitamin D status in older cerebral small vessel disease (SVD) patients. METHODS We measured serum 25(OH)D, pro-and anti-inflammatory cytokines, and mRNA levels of the vitamin D-activating enzyme, CYP27B1, as well as NF-kB, COX-2, the chemokine-CCL2, IL-1β, IL-6, TNF-α, TGF-β, and IL-10, in cerebral SVD patients aged ≥60 years presenting with vascular dementia and age and gender-matched healthy controls. RESULTS Low vitamin D status (insufficiency: serum 25(OH)D 12-20 ng/ml; deficiency: ≤12 ng/ml) was more prevalent among patients compared to controls. The mRNA levels of NF-kB, COX-2, CCL2, IL-1β, and IL-6, and serum levels of pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, and TNF-α) were significantly higher in cases compared to controls. There was a significant correlation between CYP27B1 and NF-kB, COX-2, CCL2, and IL-1β gene expression. Serum IL-1α, IL-1β, and IL-6 concentrations and the expression of CCL-2, NF-kB2, and NF-kB3 genes were higher in vitamin D-deficient subjects compared to vitamin D-sufficient subjects. There was a significant negative correlation between serum 25(OH)D and IL-1α, IL-6, and TNF-α, and a positive correlation between 25(OH)D and IL-10. CONCLUSION Low vitamin D is associated with an inflammatory response via NF-kB signaling, which could play a role in the etio-pathogenesis of SVD. Further large-scale studies are required to validate our findings.
Collapse
|
29
|
Yang X, Xu L, Zhao H, Xie T, Wang J, Wang L, Yang J. Curcumin protects against cerebral ischemia-reperfusion injury in rats by attenuating oxidative stress and inflammation: a meta-analysis and mechanism exploration. Nutr Res 2023; 113:14-28. [PMID: 36996692 DOI: 10.1016/j.nutres.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/11/2023] [Accepted: 02/26/2023] [Indexed: 03/09/2023]
Abstract
Accumulating evidence has suggested that curcumin may protect against cerebral ischemia-reperfusion injury (CIRI). However, biological mechanisms vary across studies, limiting the clinical applicability of these findings. We performed a meta-analysis on publications evaluating curcumin administration in rat models of CIRI. Furthermore, we sought to test the hypothesis that curcumin alleviates CIRI through diminishing oxidation and inflammation. We searched PubMed, Embase, Web of Science, and Cochrane from the starting date of each database to May 2022 for experimental rat studies exploring the use of curcumin after ischemia reperfusion. Included articles were assessed for bias using SYRCLE's risk of bias tool. Data were aggregated by a random effects model. Curcumin administration significantly reduced neurological deficit score (20 studies; pooled mean difference [MD] = -1.57; 95% CI, -1.78 to -1.36, P < .00001), infarct volume (18 studies; pooled MD = -17.56%; 95% CI, -20.92% to -14.20%; P < 0.00001), and brain water content (8 studies, pooled MD = -11.29%, 95% CI: -16.48%, -6.11%, P < .00001). Compared with control, the levels of superoxide dismutase, glutathione, and glutathione peroxidase were significantly higher, whereas the levels of reactive oxygen species, malondialdehyde, interleukin-1β, interleukin-6, interleukin-8, and nuclear factor kappa B were significantly lower (P < .05). Subgroup analysis raised the possibility that intervention affections differed by curcumin's dose. To our knowledge, this is the first meta-analysis of curcumin's neuroprotection and mechanisms in rat CIRI models. Our analysis suggests the neuroprotective potential of curcumin in CIRI via antioxidant activity and anti-inflammatory effect. More research is required to further confirm the effectiveness and safety of curcumin on ischemic stroke therapy.
Collapse
Affiliation(s)
- Xuyi Yang
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Liang Xu
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Hui Zhao
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Tinghui Xie
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Jiabing Wang
- Department of Pharmacy, Taizhou Municipal Hospital, Taizhou, China
| | - Lei Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianwei Yang
- General Practice, Zhejiang Taizhou Hospital, Linhai, China.
| |
Collapse
|
30
|
Wang J, Cheng C, Liu Z, Lin Y, Yang L, Zhang Z, Sun X, Zhou M, Jing P, Zhong Z. Inhibition of A1 Astrocytes and Activation of A2 Astrocytes for the Treatment of Spinal Cord Injury. Neurochem Res 2023; 48:767-780. [PMID: 36418652 DOI: 10.1007/s11064-022-03820-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/20/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
Spinal cord injury (SCI) is a serious injury to the central nervous system that causes significant physical and psychological trauma to the patient. SCI includes primary spinal cord injuries and secondary spinal cord injuries. The secondary injury refers to the pathological process or reaction after the primary injury. Although SCI has always been thought to be an incurable injury, the human nerve has the ability to repair itself after an injury. However, the reparability is limited because glial scar formation impedes functional recovery. There is a type of astrocyte that can differentiate into two forms of reactive astrocytes known as 'A1' and 'A2' astrocytes. A1 astrocytes release cytotoxic chemicals that cause neurons and oligodendrocytes to die and perform a harmful role. A2 astrocytes can produce neurotrophic factors and act as neuroprotectors. This article discusses ways to block A1 astrocytes while stimulating A2 astrocytes to formulate a new treatment for spinal cord injury.
Collapse
Affiliation(s)
- Jingxuan Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Cai Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhongbing Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yan Lin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lingling Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zijun Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoduan Sun
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Pei Jing
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
31
|
Okazaki K, Nakamura S, Koyano K, Konishi Y, Kondo M, Kusaka T. Neonatal asphyxia as an inflammatory disease: Reactive oxygen species and cytokines. Front Pediatr 2023; 11:1070743. [PMID: 36776908 PMCID: PMC9911547 DOI: 10.3389/fped.2023.1070743] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Neonatologists resuscitate asphyxiated neonates by every available means, including positive ventilation, oxygen therapy, and drugs. Asphyxiated neonates sometimes present symptoms that mimic those of inflammation, such as fever and edema. The main pathophysiology of the asphyxia is inflammation caused by hypoxic-ischemic reperfusion. At birth or in the perinatal period, neonates may suffer several, hypoxic insults, which can activate inflammatory cells and inflammatory mediator production leading to the release of larger quantities of reactive oxygen species (ROS). This in turn triggers the production of oxygen stress-induced high mobility group box-1 (HMGB-1), an endogenous damage-associated molecular patterns (DAMPs) protein bound to toll-like receptor (TLR) -4, which activates nuclear factor-kappa B (NF-κB), resulting in the production of excess inflammatory mediators. ROS and inflammatory mediators are produced not only in activated inflammatory cells but also in non-immune cells, such as endothelial cells. Hypothermia inhibits pro-inflammatory mediators. A combination therapy of hypothermia and medications, such as erythropoietin and melatonin, is attracting attention now. These medications have both anti-oxidant and anti-inflammatory effects. As the inflammatory response and oxidative stress play a critical role in the pathophysiology of neonatal asphyxia, these drugs may contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Kaoru Okazaki
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masatoshi Kondo
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
32
|
Sheng S, Ma Y, Zou Y, Hu F, Chen L. Protective effects of blocking PD-1 pathway on retinal ganglion cells in a mouse model of chronic ocular hypertension. Front Immunol 2023; 13:1094132. [PMID: 36741384 PMCID: PMC9889850 DOI: 10.3389/fimmu.2022.1094132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023] Open
Abstract
Purpose In this study, we aimed to investigate whether Programmed cell death 1 ligand 1/programmed cell death 1 ligand 2 (PD-L1/PD-L2) double knockout (dKO) has a protective effect on RGCs in a mouse model of chronic ocular hypertension (COHT). Methods We used superparamagnetic iron oxide to induce COHT in mice. Apoptosis of retinal ganglion cells (RGCs) and activation of microglia were evaluated using western blotting (WB) and immunofluorescence staining of the mouse retina. In addition, we also conducted transcriptome sequencing and further gene expression analyses using the gene ontology (GO) and Kyoto Encyclopedia of Genes (KEGG) database. Results In the mouse model of COHT, PD-L1/PD-L2 prevented the apoptosis of RGCs to some extent. Blocking the programmed cell death 1 (PD-1) pathway also increased the number of anti-inflammatory M2-activated microglia and enhanced the phosphorylation of its related pathway signal transducer and activator of transcription (STAT)6. Sequencing results showed that this protective effect may have been achieved by regulating the NF-B, tumour necrosis factor (TNF), PI3K/Akt and toll-like receptor signaling pathway etc. Conclusion Blocking the PD-1 pathway has a protective effect on RGCs in the mouse model of COHT induced by superparamagnetic iron oxide.
Collapse
Affiliation(s)
- Siqi Sheng
- Department of Ophthalmology & Vision Science, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China,Key National Health Coucil (NHC) Key Laboratory of Myopia, Fudan University, Shanghai, China,Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Yixian Ma
- Department of Ophthalmology & Vision Science, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China,Key National Health Coucil (NHC) Key Laboratory of Myopia, Fudan University, Shanghai, China,Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Yue Zou
- Department of Ophthalmology & Vision Science, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China,Key National Health Coucil (NHC) Key Laboratory of Myopia, Fudan University, Shanghai, China,Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Fangyuan Hu
- Department of Ophthalmology & Vision Science, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China,Key National Health Coucil (NHC) Key Laboratory of Myopia, Fudan University, Shanghai, China,Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ling Chen
- Department of Ophthalmology & Vision Science, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China,Key National Health Coucil (NHC) Key Laboratory of Myopia, Fudan University, Shanghai, China,Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & Ears, Nose and Throat (ENT) Hospital, Shanghai Medical School, Fudan University, Shanghai, China,*Correspondence: Ling Chen,
| |
Collapse
|
33
|
N-Acetylcysteine Administration Attenuates Sensorimotor Impairments Following Neonatal Hypoxic-Ischemic Brain Injury in Rats. Int J Mol Sci 2022; 23:ijms232416175. [PMID: 36555816 PMCID: PMC9783020 DOI: 10.3390/ijms232416175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypoxic ischemic (HI) brain injury that occurs during neonatal period has been correlated with severe neuronal damage, behavioral deficits and infant mortality. Previous evidence indicates that N-acetylcysteine (NAC), a compound with antioxidant action, exerts a potential neuroprotective effect in various neurological disorders including injury induced by brain ischemia. The aim of the present study was to investigate the role of NAC as a potential therapeutic agent in a rat model of neonatal HI brain injury and explore its long-term behavioral effects. To this end, NAC (50 mg/kg/dose, i.p.) was administered prior to and instantly after HI, in order to evaluate hippocampal and cerebral cortex damage as well as long-term functional outcome. Immunohistochemistry was used to detect inducible nitric oxide synthase (iNOS) expression. The results revealed that NAC significantly alleviated sensorimotor deficits and this effect was maintained up to adulthood. These improvements in functional outcome were associated with a significant decrease in the severity of brain damage. Moreover, NAC decreased the short-term expression of iNOS, a finding implying that iNOS activity may be suppressed and that through this action NAC may exert its therapeutic action against neonatal HI brain injury.
Collapse
|
34
|
Li Z, Zhao M, Zhang X, Lu Y, Yang Y, Xie Y, Zou Z, Zhou L, Shang R, Zhang L, Jiang F, Du D, Zhou P. TJ-M2010-5, a novel CNS drug candidate, attenuates acute cerebral ischemia-reperfusion injury through the MyD88/NF-κB and ERK pathway. Front Pharmacol 2022; 13:1080438. [PMID: 36588708 PMCID: PMC9797592 DOI: 10.3389/fphar.2022.1080438] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Cerebral ischemia-reperfusion injury (CIRI) inevitably occurs after vascular recanalization treatment for ischemic stroke. The accompanying inflammatory cascades have a major impact on outcome and regeneration after ischemic stroke. Evidences have demonstrated that TLR/MyD88/NF-κB signaling contributes to CIRI. This study aimed to investigate the druggability of MyD88 in the central nervous system (CNS) and the neuroprotective and anti-neuroinflammatory effects of the MyD88 inhibitor TJ-M2010-5 on CIRI. Methods: A middle cerebral artery occlusion (MCAO) model was used to simulate CIRI in mice. BV-2 cells were stimulated with oxygen glucose deprivation/reoxygenation (OGD/R) or lipopolysaccharide, and SH-SY5Y cells were induced by OGD/R in vitro. Neurological deficit scores and cerebral infarction volumes were evaluated. Immunofluorescence staining was performed to measure neuronal damage and apoptosis in the brain. The anti-neuroinflammatory effect of TJ-M2010-5 was evaluated by analyzing the expression of inflammatory cytokines, activation of microglia, and infiltration of peripheral myeloid cells. The expression of proteins of the MyD88/NF-κB and ERK pathway was detected by Simple Western. The concentrations of TJ-M2010-5 in the blood and brain were analyzed by liquid chromatography-mass spectrometry. Results: The cerebral infarction volume decreased in mice treated with TJ-M2010-5, with the most prominent decrease being approximately 80% of the original infarction volume. Neuronal loss and apoptosis were reduced following TJ-M2010-5 treatment. TJ-M2010-5 inhibited the infiltration of peripheral myeloid cells and the activation of microglia. TJ-M2010-5 also downregulated the expression of inflammatory cytokines and inhibited the MyD88/NF-κB and ERK pathway. Furthermore, TJ-M2010-5 showed good blood-brain barrier permeability and no neurotoxicity. Conclusion: TJ-M2010-5 has an excellent therapeutic effect on CIRI as a novel CNS drug candidate by inhibiting excessive neuroinflammatory responses.
Collapse
Affiliation(s)
- Zeyang Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Minghui Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiran Lu
- Wuhan Yangtze International School, Wuhan International Educational Center, Wuhan, China
| | - Yang Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yalong Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhimiao Zou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Liang Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Runshi Shang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Limin Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Fengchao Jiang
- Academy of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dunfeng Du
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China,*Correspondence: Dunfeng Du, dudunfeng@163; Ping Zhou,
| | - Ping Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China,*Correspondence: Dunfeng Du, dudunfeng@163; Ping Zhou,
| |
Collapse
|
35
|
Li T, Li L, Peng R, Hao H, Zhang H, Gao Y, Wang C, Li F, Liu X, Chen F, Zhang S, Zhang J. Abrocitinib Attenuates Microglia-Mediated Neuroinflammation after Traumatic Brain Injury via Inhibiting the JAK1/STAT1/NF-κB Pathway. Cells 2022; 11:cells11223588. [PMID: 36429017 PMCID: PMC9688110 DOI: 10.3390/cells11223588] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuroinflammation has been shown to play a critical role in secondary craniocerebral injury, leading to poor outcomes for TBI patients. Abrocitinib, a Janus kinase1 (JAK1) selective inhibitor approved to treat atopic dermatitis (AD) by the Food and Drug Administration (FDA), possesses a novel anti-inflammatory effect. In this study, we investigated whether abrocitinib could ameliorate neuroinflammation and exert a neuroprotective effect in traumatic brain injury (TBI) models. METHODS First, next-generation sequencing (NGS) was used to select genes closely related to neuroinflammation after TBI. Then, magnetic resonance imaging (MRI) was used to dynamically observe the changes in traumatic focus on the 1st, 3rd, and 7th days after the induction of fluid percussion injury (FPI). Moreover, abrocitinib's effects on neurobehaviors were evaluated. A routine peripheral blood test was carried out and Evans blue dye extravasation, cerebral cortical blood flow, the levels of inflammatory cytokines, and changes in the numbers of inflammatory cells were evaluated to investigate the function of abrocitinib on the 1st day post-injury. Furthermore, the JAK1/signal transducer and activator of transcription1 (STAT1)/nuclear factor kappa (NF-κB) pathway was assessed. RESULTS In vivo, abrocitinib treatment was found to shrink the trauma lesions. Compared to the TBI group, the abrocitinib treatment group showed better neurological function, less blood-brain barrier (BBB) leakage, improved intracranial blood flow, relieved inflammatory cell infiltration, and reduced levels of inflammatory cytokines. In vitro, abrocitinib treatment was shown to reduce the pro-inflammatory M1 microglia phenotype and shift microglial polarization toward the anti-inflammatory M2 phenotype. The WB and IHC results showed that abrocitinib played a neuroprotective role by restraining JAK1/STAT1/NF-κB levels after TBI. CONCLUSIONS Collectively, abrocitinib treatment after TBI is accompanied by improvements in neurological function consistent with radiological, histopathological, and biochemical changes. Therefore, abrocitinib can indeed reduce excessive neuroinflammation by restraining the JAK1/STAT1/NF-κB pathway.
Collapse
Affiliation(s)
- Tuo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Lei Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Ruilong Peng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Hongying Hao
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300000, China
- Department of Neurology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Hejun Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Department of Neurosurgery, First Hospital of Qinhuangdao, Qinhuangdao 066000, China
| | - Yalong Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Cong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Fanjian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Xilei Liu
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Correspondence: (S.Z.); (J.Z.)
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Correspondence: (S.Z.); (J.Z.)
| |
Collapse
|
36
|
Xu H, You M, Xiang X, Zhao J, Yuan P, Chu L, Xie C. Molecular Mechanism of Epimedium Extract against Ischemic Stroke Based on Network Pharmacology and Experimental Validation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3858314. [PMID: 36338345 PMCID: PMC9633197 DOI: 10.1155/2022/3858314] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2024]
Abstract
Ischemic stroke exhibits high morbidity, disability, and mortality, and treatments for ischemic stroke are limited despite intensive research. The potent neuroprotective benefits of Epimedium against ischemic stroke have gained lots of interest. Nevertheless, systematic research on the direct role and mechanisms of Epimedium in ischemic stroke is still lacking. Network pharmacology analysis coupled with experimental verification was utilized to systematically evaluate the potential pharmacological mechanism of Epimedium against ischemic stroke. The TCMSP database was used to mine the bioactive ingredients and Epimedium's targets. The DrugBank, OMIM, and GeneCards databases were employed to identify potential targets of ischemic stroke. GO and KEGG pathway analyses were also carried out. The interaction between active components and hub targets was confirmed via molecular docking. An experimental ischemic stroke model was used to evaluate the possible therapeutic mechanism of Epimedium. As a result, 23 bioactive compounds of Epimedium were selected, and 30 hub targets of Epimedium in its function against ischemic stroke were identified, and molecular docking results demonstrated good binding. The IL-17 signaling pathway was revealed as a potentially significant pathway, with the NF-κB and MAPK/ERK signaling pathways being involved. Furthermore, in vivo experiments demonstrated that Epimedium treatment could improve neurological function and reduce infarct volume. Additionally, Epimedium reduced the activation of microglia and astrocytes in both the ischemic penumbra of the hippocampus and cerebral cortex following ischemic stroke. Western blot and RT-qPCR analyses demonstrated that Epimedium not only depressed the expression of IL-1β, TNF-α, IL-6, and IL-4 but also inhibited the NF-κB and MAPK/ERK signaling pathways. This study applied network pharmacology and in vivo experiment to explore possible mechanism of Epimedium's role against ischemic stroke, which provides insight into the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongbei Xu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Mingyao You
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Xiang Xiang
- Neurosurgery Department of Chongqing University, Three Gorges Hospital, Chongqing 400010, China
| | - Jun Zhao
- Department of Neurosurgery, Dazhou Hospital of Integrated Traditional and Western Medicine, 635000, China
| | - Ping Yuan
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Lan Chu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Chenchen Xie
- Department of Neurology, Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu 610081, China
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
37
|
Senkyunolide H Affects Cerebral Ischemic Injury through Regulation on Autophagy of Neuronal Cells via P13K/AKT/mTOR Signaling Pathway. DISEASE MARKERS 2022; 2022:9211621. [PMID: 36225195 PMCID: PMC9550497 DOI: 10.1155/2022/9211621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022]
Abstract
Cerebral ischemia (CI) is associated with high global incidence and risk; therefore, its rapid and reliable therapeutic management is essential for protecting patients' lives and improving health. Senkyunolide H (SH) is remarkably effective against phlebosclerosis, oxidation, and apoptosis. Blood-brain barrier is the main obstacle impeding the delivery of drugs and xenobiotics to brain areas. Drugs' loading in nanoparticles can overcome the blood-brain barrier obstacle and thus directly and completely act on brain tissue, and such a loading can also change the half-life of drugs in vivo and lower the dosage requirement of drugs. In this study, we loaded the SH in lipid nanoparticles to improve its delivery to the brain for the therapy of CI. Thus, this study preliminarily analyzed the mechanism of SH-loaded nanoparticles in CI. The SH-loaded lipid nanoparticles were prepared and characterized with electron microscopy and PS potentiometery. The SH-loaded nanoparticles were intraperitoneally administered to CI-induced rats and brain tissue water content, and neuronal apoptosis and autophagy-associated proteins were determined. Our assays revealed SH-loaded nanoparticle's ability to reduce nerve injury and brain tissue water content in rats with CI and inhibit the apoptosis and autophagy of their neuronal cells (NCs). Additionally, under intervention with SH-loaded nanoparticles, P13K/AKT/mTOR pathway-associated proteins in brain tissue of rats decreased. As the assay results showed, SH-loaded nanoparticles can suppress the autophagy of NCs through medicating P13K/AKT/mTOR pathway and lower apoptosis, thus delivering the effect of treating CI. Results of this study indicate SH-loaded nanoparticles as promising strategy for delivery SH to brain areas for treating CI.
Collapse
|
38
|
Guo H, Zhang W, Wang Z, Li Z, Zhou J, Yang Z. Dexmedetomidine post-conditioning protects blood-brain barrier integrity by modulating microglia/macrophage polarization via inhibiting NF-κB signaling pathway in intracerebral hemorrhage. Front Mol Neurosci 2022; 15:977941. [PMID: 36172260 PMCID: PMC9512049 DOI: 10.3389/fnmol.2022.977941] [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: 06/25/2022] [Accepted: 08/10/2022] [Indexed: 11/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most devastating forms of stroke. Dexmedetomidine (DEX) has shown certain neuroprotective roles in ICH. Nevertheless, the details concerning the underlying molecular mechanism of DEX’s protective effects still need further elucidation. Herein, a model of ICH was established. The rats were randomly divided into the sham group, the ICH group, and the ICH + DEX group. Neurological outcomes, neuronal injury, and apoptosis were evaluated. Brain water content, Evans blue extravasation, and the expression of tight junction-associated proteins were also detected to assess the blood-brain barrier (BBB) integrity. Subsequently, the microglia/macrophage polarization state and inflammatory cytokine levels were observed. To further explore the underlying mechanism, NF-κB signaling pathway-associated proteins were detected. The results showed that DEX exerted neuroprotective effects against ICH-induced neurological deficits. DEX significantly increased the numbers of the surviving neurons and ameliorated neuronal cell loss and apoptosis in ICH. The rats that received the DEX displayed a lower level of brain water content and EB extravasation, moreover, ZO-1, occludin, and claudin-5 were markedly increased by DEX. Additionally, DEX facilitated M2 microglia/macrophage polarization, the M1-associated markers were reduced by DEX, while the M2-associated identification significantly increased. We found that DEX dramatically diminished pro-inflammatory cytokines expression, simultaneously promoting anti-inflammatory cytokines expression. DEX inhibited nuclear translocation of NF-κB in ICH rats. Our data suggest that DEX post-conditioning protects BBB integrity by modulating microglia/macrophage polarization via inhibiting the NF-κB signaling pathway in ICH.
Collapse
Affiliation(s)
- Hao Guo
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China.,The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Weiwei Zhang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Zhi Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Zhishan Li
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Jing Zhou
- Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Zhaoyu Yang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Institute of Integrative Medicine, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Yang X, Yun Y, Wang P, Zhao J, Sun X. Upregulation of RCAN1.4 by HIF1α alleviates OGD-induced inflammatory response in astrocytes. Ann Clin Transl Neurol 2022; 9:1224-1240. [PMID: 35836352 PMCID: PMC9380140 DOI: 10.1002/acn3.51624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/03/2022] [Accepted: 06/28/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Ischemic stroke is a leading cause of human mortality and long-term disability worldwide. As one of the main forms of regulator of calcineurin 1 (RCAN1), the contribution of RCAN1.4 in diverse biological and pathological conditions has been implicated. But the role of RCAN1.4 in ischemic stroke progression remains elusive. This study is to explore the expression changes and roles of RCAN1.4 in ischemic stroke as well as the underlying mechanisms for these changes and effects of RCAN1.4 in ischemic stroke. METHODS Middle cerebral artery occlusion model in C57BL/6J mice and oxygen-glucose deprivation (OGD) model in primary astrocytes were performed to induce the cerebral ischemic stroke. The expression pattern of RCAN1.4 was assessed using real-time quantitative PCR and western blotting in vivo and in vitro. Mechanistically, the underlying mechanism for the elevation of RCAN1.4 in the upstream was investigated. Lentiviruses were administrated, and the effect of RCAN1.4 in postischemic inflammation was clearly clarified. RESULTS Here we uncovered that RCAN1.4 was dramatically increased in mouse ischemic brains and OGD-induced primary astrocytes. HIF1α, activated upon OGD, significantly upregulated RCAN1.4 gene expression through specifically binding to the RCAN1.4 promoter region and activating its promoter activity. The functional hypoxia-responsive element (HRE) was located between -254 and -245 bp in the RCAN1.4 promoter region. Moreover, elevated RCAN1.4 alleviated the release of pro-inflammatory cytokines TNFα, IL1β, IL6 and reduced expression of iNOS, COX2 in primary astrocytes upon OGD, whereas RCAN1.4 silencing has the opposite effect. Of note, RCAN1.4 overexpression inhibited OGD-induced NF-κB activation in primary astrocytes, leading to decreased degradation of IκBα and reduced nuclear translocation of NF-κB/p65. INTERPRETATION Our results reveal a novel mechanism underscoring the upregulation of RCAN1.4 by HIF1α and the protective effect of RCAN1.4 against postischemic inflammation, suggesting its significance as a promising therapeutic target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Xiaxin Yang
- Department of NeurologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Yan Yun
- Department of RadiologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Pin Wang
- NHC Key Laboratory of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Juan Zhao
- NHC Key Laboratory of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Xiulian Sun
- NHC Key Laboratory of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- Brain Research InstituteQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health CommissionQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| |
Collapse
|
40
|
Gupta S, Guleria RS. Involvement of Nuclear Factor-κB in Inflammation and Neuronal Plasticity Associated with Post-Traumatic Stress Disorder. Cells 2022; 11:cells11132034. [PMID: 35805118 PMCID: PMC9265339 DOI: 10.3390/cells11132034] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating psychiatric condition which develops either due to stress or witnessing a traumatic situation. PTSD is characterized by acute and chronic stress response exhibit anxiety, fear, and an increased inflammatory etiology. Inflammation contributes a critical role in several parts of the brain that control fear and flashback cognatic function. It is known that impairment of the neurological circuit leads to the development of PTSD. Evidence has suggested that dysregulation of the sympathetic nervous system and hypothalamic-pituitary adrenal (HPA) axis and inflammatory responsiveness are pivotal and a greater risk in PTSD. NF-κB, a master regulator for inflammation, has been showed to modulate memory reconsolidation and synaptic plasticity; however, NF-κB’s association with PTSD remain elusive. In this review, we provide relevant findings regarding NF-κB activity in various components of brain and describe a potential mechanism linking PTSD using preclinical and clinical models. We envisage NF-κB signaling as a crucial mediator for inflammation, cognitive function, memory restoration and behavioral actions of stress and suggest that it could be used for therapeutic intervention in PTSD.
Collapse
|
41
|
Bhuiyan MIH, Young CB, Jahan I, Hasan MN, Fischer S, Meor Azlan NF, Liu M, Chattopadhyay A, Huang H, Kahle KT, Zhang J, Poloyac SM, Molyneaux BJ, Straub AC, Deng X, Gomez D, Sun D. NF-κB Signaling-Mediated Activation of WNK-SPAK-NKCC1 Cascade in Worsened Stroke Outcomes of Ang II-Hypertensive Mice. Stroke 2022; 53:1720-1734. [PMID: 35272484 PMCID: PMC9038703 DOI: 10.1161/strokeaha.121.038351] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/31/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Worsened stroke outcomes with hypertension comorbidity are insensitive to blood pressure-lowering therapies. In an experimental stroke model with comorbid hypertension, we investigated causal roles of ang II (angiotensin II)-mediated stimulation of the brain WNK (with no lysine [K] kinases)-SPAK (STE20/SPS1-related proline/alanine-rich kinase)-NKCC1 (Na-K-Cl cotransporter) complex in worsened outcomes. METHODS Saline- or ang II-infused C57BL/6J male mice underwent stroke induced by permanent occlusion of the distal branches of the middle cerebral artery. Mice were randomly assigned to receive either vehicle dimethyl sulfoxide/PBS (2 mL/kg body weight/day, IP), a novel SPAK inhibitor, 5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide (ZT-1a' 5 mg/kg per day, IP) or a NF-κB (nuclear factor-κB) inhibitor TAT-NBD (transactivator of transcription-NEMO-binding domain' 20 mg/kg per day, IP). Activation of brain NF-κB and WNK-SPAK-NKCC1 cascade as well as ischemic stroke outcomes were examined. RESULTS Stroke triggered a 2- to 5-fold increase of WNK (isoforms 1, 2, 4), SPAK/OSR1 (oxidative stress-responsive kinase 1), and NKCC1 protein in the ang II-infused hypertensive mouse brains at 24 hours after stroke, which was associated with increased nuclear translocation of phospho-NF-κB protein in the cortical neurons (a Pearson correlation r of 0.77, P<0.005). The upregulation of WNK-SPAK-NKCC1 cascade proteins resulted from increased NF-κB recruitment on Wnk1, Wnk2, Wnk4, Spak, and Nkcc1 gene promoters and was attenuated by NF-κB inhibitor TAT-NBD. Poststroke administration of SPAK inhibitor ZT-1a significantly reduced WNK-SPAK-NKCC1 complex activation, brain lesion size, and neurological function deficits in the ang II-hypertensive mice without affecting blood pressure and cerebral blood flow. CONCLUSIONS The ang II-induced stimulation of NF-κB transcriptional activity upregulates brain WNK-SPAK-NKCC1 cascade and contributes to worsened ischemic stroke outcomes, illustrating the brain WNK-SPAK-NKCC1 complex as a therapeutic target for stroke with comorbid hypertension.
Collapse
Affiliation(s)
- Mohammad Iqbal H Bhuiyan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational, and Clinical Center, PA (M.I.H.B.' D.S.)
| | - Cullen B Young
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Israt Jahan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Md Nabiul Hasan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Sydney Fischer
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom (N.F.M.A., J.Z.)
| | - Mingjun Liu
- Medicine (M.L., D.G.), University of Pittsburgh, PA
| | - Ansuman Chattopadhyay
- Molecular Biology-Information Service, Health Sciences Library System (A.C.), University of Pittsburgh, PA
| | - Huachen Huang
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston (K.T.K.)
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom (N.F.M.A., J.Z.)
| | | | - Bradley J Molyneaux
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Adam C Straub
- Pharmacology and Chemical Biology (A.C.S), University of Pittsburgh, PA
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (A.C.S., D.G.), University of Pittsburgh, PA
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China (X.D.)
| | - Delphine Gomez
- Medicine (M.L., D.G.), University of Pittsburgh, PA
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (A.C.S., D.G.), University of Pittsburgh, PA
| | - Dandan Sun
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational, and Clinical Center, PA (M.I.H.B.' D.S.)
| |
Collapse
|
42
|
Kim W, Kwon HJ, Jung HY, Hahn KR, Yoon YS, Hwang IK, Choi SY, Kim DW. Neuroprotective Effects of Purpurin Against Ischemic Damage via MAPKs, Bax, and Oxidative Stress Cascades in the Gerbil Hippocampus. Mol Neurobiol 2022; 59:2580-2592. [PMID: 35094304 PMCID: PMC9016019 DOI: 10.1007/s12035-021-02642-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/07/2021] [Indexed: 11/24/2022]
Abstract
Purpurin has various effects, including anti-inflammatory effects, and can efficiently cross the blood-brain barrier. In the present study, we investigated the effects of purpurin on oxidative stress in HT22 cells and mild brain damage in the gerbil hippocampal CA1 region induced by transient forebrain ischemia. Oxidative stress induced by H2O2 was significantly ameliorated by treatment with purpurin, based on changes in cell death, DNA fragmentation, formation of reactive oxygen species, and pro-apoptotic (Bax)/anti-apoptotic (Bcl-2) protein levels. In addition, treatment with purpurin significantly reduced the phosphorylation of c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase 1/2 (ERK), and p38 signaling in HT22 cells. Transient forebrain ischemia in gerbils led to a significant increase in locomotor activity 1 day after ischemia and significant decrease in number of surviving cells in the CA1 region 4 days after ischemia. Administration of purpurin reduced the travel distance 1 day after ischemia and abrogates the neuronal death in the hippocampal CA1 region 4 days after ischemia based on immunohistochemical and histochemical staining for NeuN and Fluoro-Jade C, respectively. Purpurin treatment significantly decreased the activation of microglia and astrocytes as well as the increases of nuclear factor kappa-light-chain-enhancer of activated B cells p65 in the hippocampal CA1 region 4 days after ischemia and ameliorated the ischemia-induced transient increases of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in the hippocampus 6 h after ischemia. In addition, purpurin significantly alleviated the ischemia-induced phosphorylation of JNK, ERK, and p38 in the hippocampus 1 day after ischemia. Furthermore, purpurin treatment significantly mitigated the increases of Bax in the hippocampus 1 day after ischemia and the lipid peroxidation based on malondialdehyde and hydroperoxides levels 2 days after ischemia. These results suggest that purpurin can be one of the potential candidates to reduce neuronal damage and inflammatory responses after oxidative stress in HT22 cells or ischemic damage in gerbils.
Collapse
Affiliation(s)
- Woosuk Kim
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
- Department of Anatomy, College of Veterinary Medicine, and Veterinary Science Research Institute, Konkuk University, Seoul, 05030, South Korea
| | - Hyun Jung Kwon
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Soo Young Choi
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea.
| |
Collapse
|
43
|
Xu T, Wang X, Ma C, Ji J, Xu W, Shao Q, Liao X, Li Y, Cheng F, Wang Q. Identification of potential regulating effect of baicalin on NFκB/CCL2/CCR2 signaling pathway in rats with cerebral ischemia by antibody-based array and bioinformatics analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114773. [PMID: 34699947 DOI: 10.1016/j.jep.2021.114773] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/10/2021] [Accepted: 10/21/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baicalin is one of the major bioactive compounds extracted from the root of Scutellaria baicalensis Georgi, which was used to treat cerebral ischemia for thounds of years. However, its biological mechanisms remains to be further explored. AIM OF THE REVIEW This study aims to identify potential biological mechanisms of baicalin against cerebral ischemia combining antibody-based array and bioinformatics analysis. METHODS A rat model of middle cerebral artery occlusion (MCAO) was constructed. Sprague-Dawley rats were randomly divided into three groups: control group, ischemic model group, and baicalin 100 mg/kg treatment group respectively. Bederson score and 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining were examined to evaluate the pharmacodynamics of baicalin treatment. Antibody-based array technology, enzyme linked immunosorbent assay (ELISA), western-blot, molecular docking, transcription factor perdiction, hematoxylin and eosin (H&E), and immunofluorescence staining were used to study the regulation of baicalin on inflammatory response after cerebral ischemia in vivo. LPS-induced RAW 264.7 macrophage inflammation model was prepared to observe the anti-inflammatory effect of baicalin in vitro. RESULTS Baicalin (100 mg/kg) reduced neurological injury score, cerebral infarction volume, and necrotic cells in MCAO rats. Baicalin inhibited the expression of CCL2, and reduced the phosphorylation levels of p65, IκBα protein and down-regulated level of CCR2. Besides, baicalin could bond to CCR2 directly, which prevented CCL2 from binding to CCR2. Furthermore, baicalin down-regulated the number of monocytes in the peripheral blood and improved the spleen index post-cerebral ischemia. In vitro, baicalin significantly inhibited the secretion of NO, IL6, TNFα, and CCL2 in macrophages and promoted the secretion of IL13, IFNG, and IL1a. CONCLUSIONS Baicalin inhibited cerebral ischemia-induced activation of the NFκB/CCL2/CCR2 pathway with multiple target effect. These data promote the therapeutic utilization of baicalin in preventing cerebral ischemia clinically.
Collapse
Affiliation(s)
- Tian Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Chongyang Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Jing Ji
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Wenxiu Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Qi Shao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Xuejing Liao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Ying Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
44
|
4R-cembranoid protects neuronal cells from oxygen-glucose deprivation by modulating microglial cell activation. Brain Res Bull 2022; 179:74-82. [PMID: 34942325 PMCID: PMC8849140 DOI: 10.1016/j.brainresbull.2021.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 02/03/2023]
Abstract
As major immune responsive cells in the central nervous system (CNS), activated microglia can present pro-inflammatory M1 phenotype aggravating the neuronal injury or anti-inflammatory M2 phenotype providing neuroprotection and promoting neuronal survival in neurodegenerative diseases. In this study, we demonstrated that a compound, 4R-cembranoid (4R, 1S, 2E, 4R, 6R,-7E, 11E-2, 7, 11-cembratriene-4, 6-diol cembranoids) promoted M2 phenotype while attenuated M1 phenotype in N9 cells, a microglial cell line. Following Lipopolysaccharides (LPS) or Oxygen-glucose deprivation (OGD) treatment, the N9 cells treated by 1 µM 4R showed an increased Arginase-1 (Arg1, a M2 marker) expression and a reduced inducible nitric oxide synthase (iNOS, M1 marker) expression. In addition, the conditioned medium of 4R-treated post-OGD N9 cells protected neuro2a cells, a neuronal cell line, from OGD-induced injury. The viability of neuro2a cells in OGD condition was increased by 54.5% after treated with the conditioned medium of 4R-treated post-OGD N9 cells. Furthermore, we demonstrated the protective mechanism of 4R was associated with a decreased TNF-α release and an increased IL-10 release from N9 cells. In conclusion, our study demonstrated that the neuroprotective effects of 4R were through the regulation of microglial activation by promoting the protective M2 activation and inhibiting the damaging M1 activation. Therefore, the findings of this study suggest that 4R could be a promising lead structure for the development of drugs for the treatment of ischemic stroke and other neurodegenerative diseases with an inflammatory component involved.
Collapse
|
45
|
Du J, Kang Z, Huang L, Zhou F, Feng X, Huang J. Protective effects of Hirudin against compartment syndrome in rabbits through the activation of Nrf2/HO-1. Injury 2022; 53:408-415. [PMID: 34872701 DOI: 10.1016/j.injury.2021.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 02/02/2023]
Abstract
Compartment syndrome generates an oxidative condition causing the death of skeletal muscle cells. Hirudin has antioxidant and anti-inflammatory properties. However, its correlation with the pathway of Nrf2/HO-1 for the protection of the skeletal muscle is unknown. We aimed to evaluate the protective efficacy of double-doses of hirudin in compartment syndrome and its association with Nrf2/HO-1 expression. Compartment syndrome was induced in rabbits and double-doses of hirudin (0-8 ATU/kg) were locally administered to select an optimal Hirudin concentration that protects skeletal muscle from damage. The tissue structural changes, W/D ratio, lipid peroxidation level by MDA assay and inflammatory factors were determined in the skeletal muscle. To determine the musculoprotective efficacy of H8 at 72 h timepoint after compartment syndrome, and its association with the Nrf2/HO-1 pathway, the following assays were performed by TUNEL assay and immunofluorescence: necrosis (high-mobility group box-1; HMGB1), Nrf2, and HO-1. In addition, the HO-1 mRNA was evaluated by qPCR. Hirudin in a dose of 8 ATU/kg (H8) presented the lowest levels of histological damage, fibrosis, W/D ratio and oxidative stress in the studied groups. Moreover, treatment with H8 markedly downregulated the level of inflammatory factors including TNF-a, IL-1β and IL-6. H8 showed a protective effect at 72 h timepoint after compartment syndrome, as revealed by a decrease in the levels of all damage markers. Nuclear translocation Nrf2 and HO-1 staining in cytoplasm were increased, and the levels of HO-1 mRNA were also increased. In conclusion, double-doses of H8 alleviate the death of muscle cells induced by oxidative stress 72 h after compartment syndrome in rabbits. This protective effect is associated with the nuclear translocation of Nrf2 and an elevated expression of HO-1.
Collapse
Affiliation(s)
- Jianhang Du
- Department of Orthopaedic, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China
| | - Zhanrong Kang
- Department of Orthopaedic, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China
| | - Libiao Huang
- Department of Orthopaedic, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China.
| | - Fei Zhou
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China
| | - Xu Feng
- Department of Orthopaedic, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China
| | - Jianming Huang
- Department of Orthopaedic, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China.
| |
Collapse
|
46
|
Dehqanizadeh B, Mohammadi ZF, Kalani AHT, Mirghani SJ. Effect of Early Exercise on Inflammatory Parameters and Apoptosis in CA1 Area of the Hippocampus Following Cerebral Ischemia-reperfusion in Rats. Brain Res Bull 2022; 182:102-110. [DOI: 10.1016/j.brainresbull.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/28/2022]
|
47
|
Huang Q, Cai G, Liu T, Liu Z. Relationships Among Gut Microbiota, Ischemic Stroke and Its Risk Factors: Based on Research Evidence. Int J Gen Med 2022; 15:2003-2023. [PMID: 35795301 PMCID: PMC9252587 DOI: 10.2147/ijgm.s353276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/08/2022] [Indexed: 11/27/2022] Open
Abstract
Stroke is a highly lethal disease and disabling illness while ischemic stroke accounts for the majority of stroke. It has been found that inflammation plays a key role in the initiation and progression of stroke, and atherosclerotic plaque rupture is considered to be the leading cause of ischemic stroke. Furthermore, chronic inflammatory diseases, such as obesity, type 2 diabetes mellitus (T2DM) and hypertension, are also considered as the high-risk factors for stroke. Recently, the topic on how gut microbiota affects human health has aroused great concern. The initiation and progression of ischemic stroke has been found to have close relation with gut microbiota dysbiosis. Hence, this manuscript briefly summarizes the roles of gut microbiota in ischemic stroke and its related risk factors, and the practicability of preventing and alleviating ischemic stroke by reconstructing gut microbiota.
Collapse
Affiliation(s)
- Qinhong Huang
- First Clinical School, Guangzhou Medical University, Guangzhou, 511436, People’s Republic of China
| | - Guannan Cai
- First Clinical School, Guangzhou Medical University, Guangzhou, 511436, People’s Republic of China
| | - Ting Liu
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, People’s Republic of China
- Correspondence: Ting Liu; Zhihua Liu, Email ;
| | - Zhihua Liu
- Department of Anorectal Surgery, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, People’s Republic of China
| |
Collapse
|
48
|
Li Y, Zhang JJ, Chen RJ, Chen L, Chen S, Yang XF, Min JW. Genistein mitigates oxidative stress and inflammation by regulating Nrf2/HO-1 and NF-κB signaling pathways in hypoxic-ischemic brain damage in neonatal mice. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:32. [PMID: 35282070 PMCID: PMC8848430 DOI: 10.21037/atm-21-4958] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/26/2021] [Indexed: 01/26/2023]
Abstract
Background Oxidative stress and neuroinflammation play crucial roles in the progression of neonatal hypoxic-ischemic brain damage (HIBD). Genistein, a natural phytoestrogen, has been found to protect against ischemic brain injury. However, its effects and potential mechanisms in HIBD have not yet been explored. Methods A neonatal mouse model of hypoxia-ischemia (HI) and a cell model of oxygen-glucose deprivation/reperfusion (OGD/R) were employed. In the in vivo study, genistein (10 mg/kg; ip) was administered in mice once daily for 3 consecutive days before the operation and once immediately after HI. The effects of genistein treatment on acute brain damage and long-term responses were evaluated. Neuronal injury and apoptosis were estimated using hematoxylin and eosin (H&E) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, respectively. The expression of apoptosis-related proteins were also measured by Western blot analysis. Dihydroethidium (DHE) staining and glutathione (GSH) and malondialdehyde (MDA) production were determined to assess the extent of oxidative stress. The messenger RNA (mRNA) levels of proinflammatory cytokines were detected using real-time quantitative polymerase chain reaction (RT-qPCR) to evaluate the extent of neuroinflammation. In the in vitro study, cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) assays, as well as propidium iodide (PI) staining, were performed to analyse the neuroprotective effects of genistein on primary cortical neurons. Western blot assays were used to detect the levels of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), phosphorylated inhibitor kappa B-α (p-IκB-α) and phosphorylated nuclear factor-kappa B (p-NF-κB) both in vivo and in vitro. Results Our results showed that genistein treatment effectively reduced cerebral infarction, attenuated neuronal injury and apoptosis, and contributed to the long-term recovery of neurological outcomes and brain atrophy in neonatal HIBD mice. Moreover, genistein ameliorated HIBD-induced oxidative stress and neuroinflammation. Meanwhile, genistein significantly increased cell viability, reversed neuronal injury and decreased cell apoptosis after OGD/R injury. Finally, the activation of the Nrf2/HO-1 pathway and inhibition of the NF-κB pathway by genistein were verified in the brain tissues of neonatal mice subjected to HIBD and in primary cortical neurons exposed to OGD/R. Conclusions Genistein exerted neuroprotective effects on HIBD by attenuating oxidative stress and neuroinflammation through the Nrf2/HO-1 and NF-κB signalling pathways.
Collapse
Affiliation(s)
- Yuan Li
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| | - Jin-Jia Zhang
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| | - Ru-Jia Chen
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| | - Ling Chen
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| | - Su Chen
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| | - Xiao-Fei Yang
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| | - Jia-Wei Min
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
49
|
Owjfard M, Bigdeli MR, Safari A, Namavar MR. Therapeutic Effects of Dimethyl Fumarate on the Rat Model of Brain Ischemia. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | | | - Mohammad Reza Namavar
- Shiraz University of Medical Sciences, Iran; Shiraz University of Medical Sciences, Iran
| |
Collapse
|
50
|
Intracellular Signaling. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|