1
|
Yao H, Tian J, Cheng S, Dou H, Zhu Y. The mechanism of hypoxia-inducible factor-1α enhancing the transcriptional activity of transferrin ferroportin 1 and regulating the Nrf2/HO-1 pathway in ferroptosis after cerebral ischemic injury. Neuroscience 2024; 559:26-38. [PMID: 39168172 DOI: 10.1016/j.neuroscience.2024.08.025] [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: 01/06/2024] [Revised: 07/05/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Cerebral ischemic/reperfusion (I/R) injury has high disability and morbidity. Hypoxia-inducible factor-1α (HIF-1α) may enhance the transcriptional activity of transferrin ferroportin 1 (FPN1) in regulating ferroptosis after cerebral ischemia injury (CII). In this study, cerebral I/R injury rat models were established and treated with pcDNA3.1-HIF-1α, pcDNA3.1-NC lentiviral plasmid, or ML385 (a specific Nrf2 inhibitor). Additionally, oxygen-glucose deprivation/reoxygenation (OGD/R) exposed PC12 cells were used as an in vitro model of cerebral ischemia and treated with pcDNA3.1-HIF-1α, si-FPN1, or ML385. The results elicited that cerebral I/R injury rats exhibited increased Longa scores, TUNEL and NeuN co-positive cells, Fe2+ concentration, ROS and HIF-1α levels, and MDA content, while reduced cell density and number, GSH content, and GPX4 protein level. Morphologically abnormal and disordered hippocampal neurons were also observed in CII rats. HIF-1α inhibited brain neuron ferroptosis and ameliorated I/R injury. HIF-1α alleviated OGD-induced PC12 cell ferroptosis. OGD/R decreased FPN1 protein level in PC12 cells, and HIF-1α enhanced FPN1 transcriptional activity. FPN1 knockdown reversed HIF-1α-mediated alleviation of OGD/R-induced ferroptosis. HIF-1α activated the Nrf2/HO-1 pathway by enhancing FPN1 expression and alleviating OGD/R-induced ferroptosis. Conjointly, HIF-1α enhanced the transcriptional activity of FPN1, activated the Nrf2/HO-1 pathway, and inhibited ferroptosis of brain neurons, thereby improving I/R injury in CII rats.
Collapse
Affiliation(s)
- Haiqian Yao
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Jianan Tian
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Shi Cheng
- Department of Orthopaedics, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Haitong Dou
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Yulan Zhu
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
2
|
Abou Taha MA, Ali FEM, Saleh IG, Akool ES. Sorafenib and edaravone protect against renal fibrosis induced by unilateral ureteral obstruction via inhibition of oxidative stress, inflammation, and RIPK-3/MLKL pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8961-8977. [PMID: 38874805 PMCID: PMC11522075 DOI: 10.1007/s00210-024-03146-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/05/2024] [Indexed: 06/15/2024]
Abstract
Renal fibrosis is the common endpoint of nearly all chronic and progressive nephropathies. Cell death and sterile inflammation are the main characteristics of renal fibrosis, which can lead to end-stage renal failure. The inflammatory reaction triggered by tissue damage is strongly related to necroptosis, a type of caspase-independent, regulated cell death. Using an animal model of unilateral ureteral obstruction (UUO), the anti-fibrotic effects of sorafenib (SOF), a multi-kinase inhibitor, and edaravone (EDV), a potent antioxidant and free radical scavenger, were examined in rats with obstructive nephropathy. Experimentally, animals were divided randomly into five groups: sham; UUO; UUO + SOF (5 mg/kg/day, P.O.); UUO + EDV (20 mg/kg/day, P.O.); and UUO + SOF + EDV groups. The kidney function biomarkers, oxidant/antioxidant status, renal mRNA expressions of TNF-α, collagen-1α, protein expressions of RIPK-1, RIPK-3, MLKL, caspase-8, HYP, MPO, and TNF-α were all significantly modulated by UUO. Administration of either SOF or EDV significantly attenuated cellular and molecular changes induced by UUO. Also, histopathological changes were improved. Moreover, SOF in combination with EDV, significantly improved UUO-induced renal fibrosis compared with each drug alone. Collectively, administration of either SOF or EDV or both of them significantly attenuated the rats with obstructive nephropathy, possibly by blocking the RIPK-3/MLKL necroptotic pathway and suppressing renal oxidative stress and inflammation.
Collapse
Affiliation(s)
- Mohamed A Abou Taha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University Assiut Branch, Assiut, 71524, Egypt
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University Assiut Branch, Assiut, 71524, Egypt.
| | - Ibrahim G Saleh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
- Department of Pharmacy Practice, Faculty of Pharmacy, Sinai University, Kantara, Ismailia, Egypt
| | - El-Sayed Akool
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Zhu X, Han X, Wang J. Sufentanil-induced Nrf2 protein ameliorates cerebral ischemia-reperfusion injury through suppressing neural ferroptosis. Int J Biol Macromol 2024; 279:135109. [PMID: 39197624 DOI: 10.1016/j.ijbiomac.2024.135109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
As an oxidative stress and inflammation-related disease, cerebral ischemia-reperfusion injury (CIRI) is a prevalent pathogenic factor of ischemic stroke (IS) and seriously degrades the life quality of human beings. As an opioid analgesic for anesthesia, Sufentanil (SUF) can activate the Nrf2 protein-induced anti-oxidant effects, which indicate that SUF may be used as alternative drug for CIRI therapy, but little is known regarding to its molecular mechanisms. Thus, this research aimed to examine whether SUF pre-treatment alleviated CIRI through the modulation of Nrf2 protein-mediated antioxidant activity. Our research revealed that middle cerebral artery occlusion/reperfusion (MCAO/R)-treated rats exhibited apparent CIRI-related symptoms and induced damages in rats' brain, which were all notably mitigated in the MCAO/R rats. The subsequent in vitro cellular experiments verified that oxygen-glucose deprivation/reoxygenation (OGD/R)-induced cytotoxicity were apparently reversed by SUF co-treatment in HT22 and BV2 cells, and it was also validated that SUF was capable of suppressing inflammation and ferroptosis in CIRI models by inhibiting oxidative stress-related damages. Mechanistically, the Akt/GSK-3β pathway was excessively activated by SUF to promote Nrf2 protein expressions and enhance Nrf2-meidated anti-oxidant effects, and it was found that SUF-induced protective effects during CIRI progression were all abrogated by co-treating cells with MK2206 (Akt inhibitor), NP-12 (GSK-3β inhibitor), or ML385 (Nrf2 inhibitor). In conclusion, SUF activated the Akt/GSK-3β pathway to initiate Nrf2 protein-mediated antioxidant effects, which further suppressed oxidative stress-related inflammation and ferroptosis to ameliorate CIRI progression, and SUF could potentially be used as novel therapeutic agent for CIRI treatment in clinic.
Collapse
Affiliation(s)
- Xuelian Zhu
- Key laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi 154000, China; Department of Anesthesiology, the First Affiliated Hospital of Jiamusi University, Jiamusi 154003, China
| | - Xi Han
- Department of Anatomy, School of Basic Medical Sciences, Jiamusi University, Jiamusi 154007, China
| | - Jingtao Wang
- Department of Anatomy, School of Basic Medical Sciences, Jiamusi University, Jiamusi 154007, China.
| |
Collapse
|
4
|
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 2024:10.1007/s12035-024-04541-6. [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] [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
|
5
|
Liao X, Huang Z, Ling H, Li W, Liu J, Lao Y, Su W. Mechanism of minocycline activating Nrf2/Hmox1 pathway to prevent ferroptosis and alleviate acute compartment syndrome. J Orthop Surg Res 2024; 19:686. [PMID: 39443986 PMCID: PMC11515506 DOI: 10.1186/s13018-024-05183-z] [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: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Acute compartment syndrome(ACS) is a perilous consequence of trauma. Acute compartment syndrome's precise cause is yet unknown. We performed studies to confirm that acute compartment syndrome can be relieved by suppressing ferroptosis and activating the Nrf2/Hmox1 pathway. METHODS We generated an ACS rat model and we conducted next-generation sequencing(NGS) of skeletal muscle tissue and identified differentially expressed target genes. Ultimately, we performed in vivo experiments to validate the presence of ferroptosis and the Nrf2/Hmox1 pathway in ACS rats. After the minocycline intervention, the drug was evaluated for its effects on ACS by examining changes associated with ferroptosis. RESULTS The bioinformatics analysis identified that the genetic changes in the disease were mostly focused on ferroptosis, with noticeable modifications in Nrf2/Hmox1. Based on the in vivo results, it was observed that ACS rats exhibited significantly elevated levels of ferroptosis compared to the control rats. The suppression of the Nrf2/Hmox1 pathway mediated by minocycline improves outcomes in ACS and reduces tissue damage after intervention. CONCLUSION Minocycline hinders ferroptosis via stimulating the Nrf2/Hmox1 pathway, which slows down the advancement of acute compartment syndrome.
Collapse
Affiliation(s)
- Xiong Liao
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
- Department of Orthopedic Trauma, The Affiliated Changsha Central Hospital (Changsha Central Hospital), Hengyang Medical School, University of South China, Changsha, Hunan, 410004, China
| | - Zhao Huang
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - He Ling
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Wencai Li
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Junjie Liu
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Yonghui Lao
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Wei Su
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China.
| |
Collapse
|
6
|
Chen Z, Li T, Tang HB, Lu ZW, Chen ZY, Zhao ZH, Yang XL, Zhao LL, Dang MJ, Li Y, Li WX, Wang XJ, Jiang PP, Zhan SQ, Zhang GL, Fan H. Edaravone Dexborneol provides neuroprotective effect by inhibiting neurotoxic activation of astrocytes through inhibiting NF-κB signaling in cortical ischemia. Brain Res Bull 2024; 218:111097. [PMID: 39395778 DOI: 10.1016/j.brainresbull.2024.111097] [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: 07/24/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
Edaravone Dexborneol (EDB), comprised of edaravone and (+)- bornel, has been demonstrated to have synergistic effects of antioxidant and anti-inflammatory, which makes it to be applied for stroke as a protectant. However, the underlying mechanism of neuroprotection of EDB has not been fully elucidated. Increasing evidence has shown that neurotoxic A1 astrocytes were closely related to neuronal death after cerebral ischemia. However, whether EDB could provide neuroprotection by modulating the activation of astrocytes has not yet been elucidated. The present study aimed to explore whether EDB afforded neuroprotection by modulating A1 polarization of astrocytes and the down-stream signaling after cerebral ischemia. We first validated the neuroprotective effects of EDB in mice suffering focal cerebral ischemia via evaluating behavioral test, infarct volumes and neuronal survival. As for the down-stream signaling, our data further showed that EDB alleviated neuronal death by suppressing activation of neurotoxic A1 astrocytes via inhibition of NF-κB signaling pathway in vitro. Additionally, administration of EDB reduced the number of A1 reactive astrocytes in mice of focal cerebral ischemia. The above findings demonstrated that EDB provided neuroprotective effect by inhibiting neurotoxic activation of A1 astrocytes in animal model of cerebral ischemia, which indicated that EDB-mediated phenotypic regulation of astrocytes is a potential research direction to promote neurological recovery in central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Tao Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Hai-Bin Tang
- Department of Laboratory Medicine, Xi'an Central Hospital, Xi'an Jiaotong University, 161 Xi Wu Road, Xi'an, Shaanxi 710003, China.
| | - Zi-Wei Lu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Zi-Yi Chen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Zhi-Hong Zhao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Xue-Ling Yang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Li-Li Zhao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Mei-Juan Dang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Ye Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Wen-Xian Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Xiao-Juan Wang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Peng-Peng Jiang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Shu-Qin Zhan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Gui-Lian Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Hong Fan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
7
|
Huang M, Mo Y, Lei H, Chen M. Edaravone: A Possible Treatment for Acute Lung Injury. Int J Gen Med 2024; 17:3975-3986. [PMID: 39286534 PMCID: PMC11403130 DOI: 10.2147/ijgm.s467891] [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: 05/28/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Despite technological advances in science and medicine, acute lung injury (ALI) is still associated with high mortality rates in the ICU. Therefore, finding novel drugs and treatment approaches is crucial to preventing ALI. Drug repurposing is a common practice in clinical research, primarily for drugs that have previously received approval for use in patients, to investigate novel uses of drugs and therapies. One such medication is edaravone, which is a highly effective free-radical scavenger that also has anti-inflammatory, anti-apoptotic, antioxidant, and anti-fibrotic effects. Both basic and clinical studies have shown that edaravone can treat different types of lung injury through its distinct properties. Edaravone exhibits significant protective benefits and holds promising clinical treatment potential for ALI caused by diverse factors, thereby offering a novel approach to treating ALI. This study aims to provide new insights and treatment options for ALI by reviewing both basic and clinical research on the use of edaravone. The focus is on evaluating the effectiveness of edaravone in treating ALI caused by various factors.
Collapse
Affiliation(s)
- Ma Huang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yalan Mo
- Centre for Infectious Diseases, General Hospital of Hunan Medical College, Huaihua, Hunan, People's Republic of China
| | - Haiyun Lei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| |
Collapse
|
8
|
Wang J, Du L, Zhang T, Chu Y, Wang Y, Wang Y, Ji X, Kang Y, Cui R, Zhang G, Liu J, Shi G. Edaravone Dexborneol ameliorates the cognitive deficits of APP/PS1 mice by inhibiting TLR4/MAPK signaling pathway via upregulating TREM2. Neuropharmacology 2024; 255:110006. [PMID: 38763325 DOI: 10.1016/j.neuropharm.2024.110006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Currently, there are no effective therapeutic agents available to treat Alzheimer's disease (AD). However, edaravone dexborneol (EDB), a novel composite agent used to treat acute ischemic stroke, has recently been shown to exert efficacious neuroprotective effects. However, whether EDB can ameliorate cognitive deficits in AD currently remains unclear. To this end, we explored the effects of EDB on AD and its potential mechanisms using an AD animal model (male APP/PS1 mice) treated with EDB for 10 weeks starting at 6 months of age. Subsequent analyses revealed that EDB-treated APP/PS1 mice exhibited improved cognitive abilities compared to untreated APP/PS1 mice. Administration of EDB in APP/PS1 mice further alleviated neuropathological alterations of the hippocampus, including Aβ deposition, pyramidal cell karyopyknosis, and oxidative damage, and significantly decreased the levels of inflammatory cytokines (IL-1β, IL-6 and TNF-α) and COX-2 in the hippocampus of APP/PS1 mice. Transcriptome sequencing analysis demonstrated the critical role of the inflammatory reaction in EDB treatment in APP/PS1 mice, indicating that the alleviation of the inflammatory reaction by EDB in the hippocampus of APP/PS1 mice was linked to the action of the TREM2/TLR4/MAPK signaling pathway. Further in vitro investigations showed that EDB suppressed neuroinflammation in LPS-stimulated BV2 cells by inhibiting the TLR4/MAPK signaling pathway and upregulating TREM2 expression. Thus, the findings of the present study demonstrate that EDB is a promising therapeutic agent for AD-related cognitive dysfunction.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China; Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Longyuan Du
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianyun Zhang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yun Chu
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yue Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaoming Ji
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yunxiao Kang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Rui Cui
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Guoliang Zhang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Junyan Liu
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Geming Shi
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
9
|
Wenzhi Y, Xiangyi L, Dongsheng F. The prion-like effect and prion-like protein targeting strategy in amyotrophic lateral sclerosis. Heliyon 2024; 10:e34963. [PMID: 39170125 PMCID: PMC11336370 DOI: 10.1016/j.heliyon.2024.e34963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Pathological proteins in amyotrophic lateral sclerosis (ALS), such as superoxide dismutase 1, TAR DNA-binding protein 43, and fused in sarcoma, exhibit a prion-like pattern. All these proteins have a low-complexity domain and seeding activity in cells. In this review, we summarize the studies on the prion-like effect of these proteins and list six prion-like protein targeting strategies that we believe have potential for ALS therapy, including antisense oligonucleotides, antibody-based technology, peptide, protein chaperone, autophagy enhancement, and heteromultivalent compounds. Considering the pathological complexity and heterogeneity of ALS, we believe that the final solution to ALS therapy is most likely to be an individualized cocktail therapy, including clearance of toxicity, blockage of pathological progress, and protection of neurons.
Collapse
Affiliation(s)
- Yang Wenzhi
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Liu Xiangyi
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Fan Dongsheng
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| |
Collapse
|
10
|
Guo H, Li H, Jia Z, Ma S, Zhang J. Edaravone dexborneol attenuates cognitive impairment in a rat model of vascular dementia by inhibiting hippocampal oxidative stress and inflammatory responses and modulating the NMDA receptor signaling pathway. Brain Res 2024; 1833:148917. [PMID: 38582415 DOI: 10.1016/j.brainres.2024.148917] [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: 02/05/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Exploring the intricate pathogenesis of Vascular Dementia (VD), there is a noted absence of potent treatments available in the current medical landscape. A new brain-protective medication developed in China, Edaravone dexboeol (EDB), has shown promise due to its antioxidant and anti-inflammatory properties, albeit with a need for additional research to elucidate its role and mechanisms in VD contexts. In a research setup, a VD model was established utilizing Sprague-Dawley (SD) rats, subjected to permanent bilateral typical carotid artery occlusion (2VO). Behavioral assessment of the rats was conducted using the Bederson test and pole climbing test, while cognitive abilities, particularly learning and memory, were evaluated via the novel object recognition test and the Morris water maze test. Ensuing, the levels of malondialdehyde (MDA), superoxide dismutase (SOD), IL-1β, IL-6, IL-4, and tumor necrosis factor-α (TNF-α) were determined through Enzyme-Linked Immunosorbent Assay (ELISA). Synaptic plasticity-related proteins, synaptophysin (SYP), post-synaptic density protein 95 (PSD-95), and N-methyl-D-aspartate (NMDA) receptor proteins (NR1, NR2A, NR2B) were investigated via Western blotting technique. The findings imply that EDB has the potential to ameliorate cognitive deficiencies, attributed to VD, by mitigating oxidative stress, dampening inflammatory responses, and modulating the NMDA receptor signaling pathway, furnishing new perspectives into EDB's mechanism and proposing potential avenues for therapeutic strategies in managing VD.
Collapse
Affiliation(s)
- Hui Guo
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Haodong Li
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhisheng Jia
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shuyu Ma
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Jin Zhang
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China.
| |
Collapse
|
11
|
Zheng H, Wu H, Wang D, Wang S, Ji D, Liu X, Gao G, Su X, Zhang Y, Ling Y. Research progress of prodrugs for the treatment of cerebral ischemia. Eur J Med Chem 2024; 272:116457. [PMID: 38704941 DOI: 10.1016/j.ejmech.2024.116457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
It is well-known that pharmacotherapy plays a pivotal role in the treatment and prevention of cerebral ischemia. Nevertheless, existing drugs, including numerous natural products, encounter various challenges when applied in cerebral ischemia treatment. These challenges comprise poor brain absorption due to low blood-brain barrier (BBB) permeability, limited water solubility, inadequate bioavailability, poor stability, and rapid metabolism. To address these issues, researchers have turned to prodrug strategies, aiming to mitigate or eliminate the adverse properties of parent drug molecules. In vivo metabolism or enzymatic reactions convert prodrugs into active parent drugs, thereby augmenting BBB permeability, improving bioavailability and stability, and reducing toxicity to normal tissues, ultimately aiming to enhance treatment efficacy and safety. This comprehensive review delves into multiple effective prodrug strategies, providing a detailed description of representative prodrugs developed over the past two decades. It underscores the potential of prodrug approaches to improve the therapeutic outcomes of currently available drugs for cerebral ischemia. The publication of this review serves to enrich current research progress on prodrug strategies for the treatment and prevention of cerebral ischemia. Furthermore, it seeks to offer valuable insights for pharmaceutical chemists in this field, offer guidance for the development of drugs for cerebral ischemia, and provide patients with safer and more effective drug treatment options.
Collapse
Affiliation(s)
- Hongwei Zheng
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Hongmei Wu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dezhi Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Sijia Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dongliang Ji
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xiao Liu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Ge Gao
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yanan Zhang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yong Ling
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| |
Collapse
|
12
|
Lin P, Lin C, Diao L. RBM3 Ameliorates Acute Brain Injury-induced Inflammation and Oxidative Stress by Stabilizing GAS6 mRNA Through Nrf2 Signaling Pathway. Neuroscience 2024; 547:74-87. [PMID: 38555015 DOI: 10.1016/j.neuroscience.2024.03.028] [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: 01/22/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
RNA-binding motif protein 3 (RBM3), as a cold-inducible protein, exhibits neuroprotective function in brain disorders. This study was conducted to investigate the effects of RBM3 on acute brain injury (ABI) and its underlying mechanism. The cerebral injury (CI) rat model and oxygen-glucose deprivation (OGD) cell model were established. The neurological severity score, wire-grip score, morris water maze test, and Y-maze test were used to detect the neurological damage, vestibular motor, learning, and memory functions. Cerebral injury, apoptosis, oxidative stress, and inflammatory level were evaluated by hematoxylin-eosin and TUNEL staining and specific kits. Flow cytometry was used to analyze the apoptosis rate. The relationship between RBM3 and growth arrest specific (GAS) 6 was analyzed by RNA immunoprecipitation assay. The results indicated that RBM3 recovered of neurological function and behaviour impairment of CI rats. Additionally, RBM3 reversed the increased oxidative stress, inflammatory level, and apoptosis induced by CI and OGD. RBM3 interacted with GAS6 to activate the Nrf2 signaling pathway, thus playing neuroprotection on ABI. Besides, the results of RBM3 treatment were similar to those of mild hypothermia treatment. In summary, RBM3 exerted neuroprotection and ameliorated inflammatory levels and oxidative stress by stabilizing GAS6 mRNA through the Nrf2 signaling pathway, suggesting that RBM3 might be a potential therapeutic candidate for treating ABI.
Collapse
Affiliation(s)
- Pingqing Lin
- Department Of Emergency, Fuzhou Second General Hospital, Fuzhou City, Fujian Province 350007, China.
| | - Chengshi Lin
- Department Of Emergency, Fuzhou Second General Hospital, Fuzhou City, Fujian Province 350007, China
| | - Liangbiao Diao
- Department Of Nephrology, Fuzhou Second General Hospital, Fuzhou City, Fujian Province 350007, China
| |
Collapse
|
13
|
Ma XX, Xie HY, Hou PP, Wang XJ, Zhou W, Wang ZH. Nuclear Factor Erythroid 2-Related Factor 2 is Essential for Low-Normobaric Oxygen Treatment-Mediated Blood-Brain Barrier Protection Following Ischemic Stroke. Mol Neurobiol 2024; 61:2938-2948. [PMID: 37950788 DOI: 10.1007/s12035-023-03767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/01/2023] [Indexed: 11/13/2023]
Abstract
Cerebral ischemia/reperfusion (I/R) injury increases blood-brain barrier (BBB) permeability, leading to hemorrhagic transformation and brain edema. Normobaric oxygen (NBO) is a routine clinical treatment strategy for this condition. However, its neuroprotective effects remain controversial. This study investigated the effect of different NBO concentrations on I/R injury and explores the involvement of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in the underlying mechanism. A mouse middle cerebral artery occlusion (MCAO) model, and an oxygen and glucose deprivation (OGD) model featuring mouse brain microvascular endothelial cells (ECs) called bEnd.3, were used to investigate the effect of NBO on I/R injury. A reactive oxygen species (ROS) inducer and Nrf2-knockdown by RNA were used to explore whether the Nrf2 pathway mediates the effect of NBO on cerebrovascular ECs. In the early stage of MCAO, 40% O2 NBO exposure significantly improved blood perfusion in the ischemic area and effectively relieved BBB permeability, cerebral edema, cerebral injury, and neurological function after MCAO. In the OGD model, 40% O2 NBO exposure significantly reduced apoptosis, inhibited ROS generation, reduced ER stress, upregulated the expression of tight junction proteins, and stabilized the permeability of ECs. Blocking the Nrf2 pathway nullified the protective effect of 40% O2 NBO on ECs after OGD. Finally, our study confirmed that low concentrations of NBO have a neuroprotective effect on I/R by activating the Nrf2 pathway in ECs.
Collapse
Affiliation(s)
- Xiao-Xiao Ma
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Yi Xie
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pin-Pin Hou
- Central Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Jing Wang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhen-Hong Wang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
D’Apolito E, Sisalli MJ, Tufano M, Annunziato L, Scorziello A. Oxidative Metabolism in Brain Ischemia and Preconditioning: Two Sides of the Same Coin. Antioxidants (Basel) 2024; 13:547. [PMID: 38790652 PMCID: PMC11117774 DOI: 10.3390/antiox13050547] [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: 03/20/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Brain ischemia is one of the major causes of chronic disability and death worldwide. It is related to insufficient blood supply to cerebral tissue, which induces irreversible or reversible intracellular effects depending on the time and intensity of the ischemic event. Indeed, neuronal function may be restored in some conditions, such as transient ischemic attack (TIA), which may be responsible for protecting against a subsequent lethal ischemic insult. It is well known that the brain requires high levels of oxygen and glucose to ensure cellular metabolism and energy production and that damage caused by oxygen impairment is tightly related to the brain's low antioxidant capacity. Oxygen is a key player in mitochondrial oxidative phosphorylation (OXPHOS), during which reactive oxygen species (ROS) synthesis can occur as a physiological side-product of the process. Indeed, besides producing adenosine triphosphate (ATP) under normal physiological conditions, mitochondria are the primary source of ROS within the cell. This is because, in 0.2-2% of cases, the escape of electrons from complex I (NADPH-dehydrogenase) and III of the electron transport chain occurring in mitochondria during ATP synthesis leads to the production of the superoxide radical anion (O2•-), which exerts detrimental intracellular effects owing to its high molecular instability. Along with ROS, reactive nitrosative species (RNS) also contribute to the production of free radicals. When the accumulation of ROS and RNS occurs, it can cause membrane lipid peroxidation and DNA damage. Here, we describe the intracellular pathways activated in brain tissue after a lethal/sub lethal ischemic event like stroke or ischemic tolerance, respectively, highlighting the important role played by oxidative stress and mitochondrial dysfunction in the onset of the two different ischemic conditions.
Collapse
Affiliation(s)
- Elena D’Apolito
- Division of Pharmacology, Department of Neuroscience Reproductive Sciences and Dentistry, Federico II University of Naples, 80131 Napoli, Italy; (E.D.); (M.T.)
| | - Maria Josè Sisalli
- Department of Translational Medicine, Federico II University of Naples, 80131 Napoli, Italy;
| | - Michele Tufano
- Division of Pharmacology, Department of Neuroscience Reproductive Sciences and Dentistry, Federico II University of Naples, 80131 Napoli, Italy; (E.D.); (M.T.)
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience Reproductive Sciences and Dentistry, Federico II University of Naples, 80131 Napoli, Italy; (E.D.); (M.T.)
| |
Collapse
|
15
|
Zhang L, Bai XY, Sun KY, Li X, Zhang ZQ, Liu YD, Xiang Y, Liu XL. A New Perspective in the Treatment of Ischemic Stroke: Ferroptosis. Neurochem Res 2024; 49:815-833. [PMID: 38170383 DOI: 10.1007/s11064-023-04096-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/14/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024]
Abstract
Ischemic stroke is a common neurological disease. Currently, there are no Food and Drug Administration-approved drugs that can maximize the improvement in ischemic stroke-induced nerve damage. Hence, treating ischemic stroke remains a clinical challenge. Ferroptosis has been increasingly studied in recent years, and it is closely related to the pathophysiological process of ischemic stroke. Iron overload, reactive oxygen species accumulation, lipid peroxidation, and glutamate accumulation associated with ferroptosis are all present in ischemic stroke. This article focuses on describing the relationship between ferroptosis and ischemic stroke and summarizes the relevant substances that ameliorate ischemic stroke-induced neurological damage by inhibiting ferroptosis. Finally, the problems in the treatment of ischemic stroke targeting ferroptosis are discussed, hoping to provide a new direction for its treatment.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Ke Yao Sun
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Xuan Li
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Zhao Qi Zhang
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Yi Ding Liu
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Xiao Long Liu
- School of Medicine, Yan'an University, Yan'an, 716000, China.
| |
Collapse
|
16
|
Liu N, Ji Y, Liu R, Jin X. The state of astragaloside IV research: A bibliometric and visualized analysis. Fundam Clin Pharmacol 2024; 38:208-224. [PMID: 37700611 DOI: 10.1111/fcp.12956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Astragaloside IV has emerged as a pharmaceutical monomer with great medical applications and potential. Astragaloside IV has many effects such as improving myocardial ischemia, cerebral ischemia-reperfusion injury, anti-inflammatory, analgesic, antiviral, promoting lymphocyte proliferation, and antitumor effects. However, there are few bibliometric studies on astragaloside IV. OBJECTIVES We aim to visualize the hotspots and trends in astragaloside IV research through bibliometric analysis to further understand the future development of basic and clinical research. Methods The articles and reviews on astragaloside IV were screened from the Web of Science Core Collection, and knowledge maps were generated using CiteSpace software. Bibliometric analysis was performed on 971 articles published from 1998 to 2022. RESULTS The number of articles on astragaloside IV increased yearly. These publications came from 42 countries/regions, with China being the largest. The primary research institutions were Shanghai University of Traditional Chinese Medicine and Guangzhou University of Traditional Chinese Medicine. Journal of Ethnopharmacology was the most studied journal and co-cited journal. A total of 473 authors were included, among which Hongxin Wang had the highest number of publications and Zhang Wd had the highest total citation frequency. After analysis, the most common keywords are astragaloside IV, expression, and oxidative stress. Cardiovascular disease, cerebral ischemia, cancer, and kidney disease are current and developing research fields. CONCLUSION This study used bibliometrics and visualization methods to analyze the research hotspots and trends of astragaloside IV. Astragaloside IV on ischemia-reperfusion injury, cancer, and tumor may become the focus of future research.
Collapse
Affiliation(s)
- Ning Liu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yansu Ji
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Rui Liu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Jin
- Military Medicine Section, Logistics University of Chinese People's Armed Police Force, Tianjin, China
| |
Collapse
|
17
|
Wang C, Gu HQ, Dong Q, Xu A, Wang N, Yang Y, Wang F, Wang Y. Rationale and design of Treatment of Acute Ischaemic Stroke with Edaravone Dexborneol II (TASTE-2): a multicentre randomised controlled trial. Stroke Vasc Neurol 2024:svn-2023-002938. [PMID: 38471696 DOI: 10.1136/svn-2023-002938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/07/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Edaravone dexborneol is believed to be a novel cytoprotective drug, demonstrating a synergistic combination of antioxidative and anti-inflammatory properties in animal models. The Treatment of Acute Ischaemic Stroke with Edaravone Dexborneol (TASTE) trial demonstrated its superior efficacy over edaravone alone for acute ischaemic stroke (AIS) patients. However, its efficacy in individuals undergoing endovascular therapy (EVT) remains uncertain. AIM To clarify the rationale and design of the TASTE II (TASTE-2) trial. DESIGN The TASTE-2 is a multicentre, double-blind, randomised, placebo-controlled trial designed to evaluate the efficacy and safety of edaravone dexborneol in patients with AIS and large-vessel occlusion in the anterior circulation. The eligible participants, presenting with a National Institute of Health Stroke Scale score between 6 and 25 (range 0-42, with larger values suggesting severe neurological dysfunction) and an Alberta Stroke Program Early Computed Tomography Score ranging from 6 to 10 (range 0-10, with smaller values suggesting larger infarction) within the initial 24 hours after symptom onset, will be randomly allocated to either the edaravone dexborneol group or the placebo group in equal proportions prior to thrombectomy. The treatment will be continuously administered for a duration of 10-14 days. A follow-up period of 90 days will be implemented for all participants. STUDY OUTCOMES The primary efficacy outcome is defined as achieving favourable functional independence, measured by a modified Rankin Scale of 0-2 at 90 days. The primary safety outcome focuses on the incidence of serious adverse events. DISCUSSION The TASTE-2 trial will provide evidence to determine whether the administration of edaravone dexborneol in AIS patients undergoing EVT could yield significant improvements in neurological function.
Collapse
Affiliation(s)
- Chunjuan Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong-Qiu Gu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
| | - Anding Xu
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ning Wang
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Yi Yang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Feng Wang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Khassafi N, Azami Tameh A, Mirzaei H, Rafat A, Barati S, Khassafi N, Vahidinia Z. Crosstalk between Nrf2 signaling pathway and inflammation in ischemic stroke: Mechanisms of action and therapeutic implications. Exp Neurol 2024; 373:114655. [PMID: 38110142 DOI: 10.1016/j.expneurol.2023.114655] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
One of the major causes of long-term disability and mortality is ischemic stroke that enjoys limited treatment approaches. On the one hand, oxidative stress, induced by excessive generation of reactive oxygen species (ROS), plays a critical role in post-stroke inflammatory response. Increased ROS generation is one of the basic factors in the progression of stroke-induced neuroinflammation. Moreover, intravenous (IV) thrombolysis using recombinant tissue plasminogen activator (rtPA) as the only medication approved for patients with acute ischemic stroke who suffer from some clinical restrictions it could not cover the complicated episodes that happen after stroke. Thus, identifying novel therapeutic targets is crucial for successful preparation of new medicines. Recent evidence indicates that the transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2) contributes significantly to regulating the antioxidant production in cytosol, which causes antiinflammatory effects on neurons. New findings have shown a relationship between activation of the Nrf2 and glial cells, nuclear factor kappa B (NF-κB) pathway, the nucleotide-binding domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signaling, and expression of inflammatory markers, suggesting induction of Nrf2 activation can represent a promising therapeutic alternative as the modulators of Nrf2 dependent pathways for targeting inflammatory responses in neural tissue. Hence, this review addresses the relationship of Nrf2 signaling with inflammation and Nrf2 activators' potential as therapeutic agents. This review helps to improve required knowledge for focused therapy and the creation of modern and improved treatment choices for patients with ischemic stroke.
Collapse
Affiliation(s)
- Negar Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Rafat
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Negin Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
19
|
Shen G, Lou C, Li Q, Zhao B, Luo Y, Wu F, Jiao D, Fang M, Geng Y. Edaravone dexborneol alleviates cerebral ischemia-reperfusion injury through NF-κB/NLRP3 signal pathway. Anat Rec (Hoboken) 2024; 307:372-384. [PMID: 37475155 DOI: 10.1002/ar.25296] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/25/2023] [Accepted: 07/08/2023] [Indexed: 07/22/2023]
Abstract
Inflammatory injury following ischemia-reperfusion (I/R) severely limits the efficacy of stroke treatment. Edaravone dexborneol (C.EDA) has been shown to reduce inflammation following a cerebral hemorrhage. However, the precise anti-inflammatory mechanism of C.EDA is unknown. In this study, we investigated whether C.EDA provides neuroprotection after I/R in rats, as well as the potential mechanisms involved. A middle cerebral artery occlusion/reperfusion (I/R) model was created using Sprague-Dawley rats. The blood flow of the central cerebral artery was monitored by a laser speckle imaging system. The neurological score was used to assess behavioral improvement. Cerebral infarction volume was measured by TTC staining. And the integrity of the blood-brain barrier was detected by Evan's blue staining. The expression of the nuclear factor kappa-B (NF-κB)/ the NOD-like receptor protein (NLRP3) inflammasome signal pathway and microglia polarization were detected by immunofluorescence and Western blotting. The cerebral blood flow ratio indicates that the cerebral I/R model was successfully established. After reperfusion for 72 h, the improvement of neurological scores, infarct volume reduction, and integrity of the blood-brain barrier was observed in I/R rats with C.EDA treatment. Meanwhile, the immunofluorescence result showed that the expression of iNOS, NLRP3, and NF-κB protein was decreased and the level of Arg1 was increased. Western blot analysis showed that the expression of NF-κB/NLRP3 signal pathway-related protein was decreased. In conclusion, this study indicates that C.EDA alleviates I/R injury by blocking the activation of the NLRP3 inflammasome and regulating the polarization of M1/M2 microglia via the NF-κB signal pathway.
Collapse
Affiliation(s)
- Guanghong Shen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chengjian Lou
- Department of Neurosurgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Qunfeng Li
- Department of Medicine, QuZhou College of Technology, Quzhou, Zhejiang, China
| | - Bingxin Zhao
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatric, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dian Jiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Marong Fang
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Xi Y, Ma J, Lu S. Favorable neuroprotective effect of intra-arterial application of edaravone dexborneol in ischemic stroke rats. J Stroke Cerebrovasc Dis 2023; 32:107356. [PMID: 37740991 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107356] [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: 03/26/2023] [Revised: 08/07/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the neuroprotective effects of intra-arterial administration of edaravone dexborneol in rats with acute ischemic stroke and determine the optimal dose. MATERIALS AND METHODS Firstly, 120 male Sprague-Dawley rats (265-300 g) were selected to establish ischemic stroke models and were randomly divided into groups of sham-operation (Sham group), cerebral ischemia-reperfusion (IS group), permanent focal ischemia (PI group) and treatment (2MG group: 2 mg/kg, 4MG group: 4 mg/kg, 6MG group: 6 mg/kg) groups. There are 20 rats in each group, and ten rats in each group were randomly selected for Longa score and 2,3,5-triphenyl tetrazolium chloride staining to observe the changes in neurological function and the proportion of cerebral infarct volume in each group. Secondly, the remaining ten rats in each group were scored for Longa and tested for free radicals (hydroxyl radical; peroxynitrite; nitric oxide) and pro-inflammatory cytokines (interleukin 6; interleukin-1β; tumor necrosis factor-α). We monitored changes in the indicators in each group of rats. RESULTS There were no significant differences among the enrolled Sprague-Dawley rats concerning age, sex, and feeding conditions. Edaravone dexborneol could significantly reduce the cerebral levels of hydroxyl radical, interleukin 6, interleukin-1β, tumor necrosis factor-α, and their behavioral scores of acute ischemic stroke rats after a single dose in the carotid artery. The results suggested that 4 mg/kg might be an appropriate dose. CONCLUSIONS A single intra-arterial administration of edaravone dexborneol can improve neurobehavioral function and alleviate cerebral injury in acute ischemic stroke rats through anti-inflammatory and free radical scavenging effects.
Collapse
Affiliation(s)
- Yalin Xi
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, China
| | - Jingxia Ma
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, China
| | - Shujun Lu
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, China.
| |
Collapse
|
21
|
Zhang J, Xiao Y, Liu H, Xu L, Guo X, Gao Y, Li M, Xu J, Qi Q, Lv P. Edaravone Dexborneol Alleviates Neuroinflammation by Reducing Neuroglial Cell Proliferation and Suppresses Neuronal Apoptosis/Autophagy in Vascular Dementia Rats. Neurochem Res 2023; 48:3113-3128. [PMID: 37338792 DOI: 10.1007/s11064-023-03973-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023]
Abstract
More and more evidence shows that the pathological mechanism of vascular dementia (VD) is closely related to oxidative stress injury, cell apoptosis, autophagy, inflammatory response, excitatory amino acid toxicity, synaptic plasticity change, calcium overload, and other processes. Edaravone dexborneol (EDB) is a new type of neuroprotective agent that can improve the neurological damage caused by an ischemic stroke. Previous studies showed that EDB has effects on synergistic antioxidants and induces anti-apoptotic responses. However, it remains unclear whether EDB can affect apoptosis and autophagy by activating the PI3K/Akt/mTOR signaling pathway and its impact on the neuroglial cells. In this study, we established the VD model of rats by bilateral carotid artery occlusion to explore the neuroprotective effect of EDB and its mechanism. Morris Water Maze test was applied to assess the cognitive function of rats. H&E and TUNEL staining were applied to observe the cellular structure of the hippocampus. Immunofluorescence labeling was used to observe the proliferation of astrocytes and microglia. ELISA was applied to examine the levels of TNF-α, IL-1β and IL-6, and RT-PCR was applied to examine their mRNA expression levels. Western blotting was applied to examine apoptosis-related proteins (Bax, Bcl-2, Caspase-3), autophagy-related proteins (Beclin-1, P62, LC3B), PI3K/Akt/mTOR signaling pathway proteins and their phosphorylation levels. The results indicated that EDB ameliorates learning and memory in rats subjected to the VD model, alleviates neuroinflammatory response by reducing the proliferation of the neuroglial cell and inhibits apoptosis and autophagy, which may be mediated by the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yining Xiao
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Hongna Liu
- Department of Geriatric Gastroenterology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Lili Xu
- Graduate School of Hebei North University, Zhangjiakou, 075000, China
| | - Xing Guo
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, 050051, China
| | - Yaran Gao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Meixi Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Jing Xu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Qianqian Qi
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China.
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China.
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China.
| |
Collapse
|
22
|
Liu P, Liu H, Wei L, Shi X, Wang W, Yan S, Zhou W, Zhang J, Han S. Docetaxel-induced cognitive impairment in rats can be ameliorated by edaravone dexborneol: Evidence from the indicators of biological behavior and anisotropic fraction. Front Neurosci 2023; 17:1167425. [PMID: 37077321 PMCID: PMC10106566 DOI: 10.3389/fnins.2023.1167425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
ObjectiveThis study aimed to investigate the effect of Edaravone Dexborneol (ED) on impaired learning and memory in docetaxel (DTX)-treated rats using cognitive behavior assessments and magnetic resonance diffusion tensor imaging (DTI).Materials and methodsIn total, 24 male Sprague–Dawley rats were divided into control, low-dose DTX (L-DTX) model, and high-dose DTX(H-DTX) model groups, with eight rats in each group, numbered 1–8. The rats were intraperitoneally injected with 1.5 mL of either normal saline (control group), or 3 mg/kg and 6 mg/kg DTX (L-DTX and H-DTX groups, respectively), once a week for 4 weeks. The learning and memory abilities of each group were tested using a water maze. At the end of the water maze test, rats 1–4 in each group were treated with ED (3 mg/kg, 1 mL), and rats 5–8 were injected with an equal volume of normal saline once a day for 2 weeks. The learning and memory abilities of each group were evaluated again using the water maze test, and the image differences in the hippocampus of each group were analyzed using DTI.Results(1) H-DTX group (32.33 ± 7.83) had the longest escape latency, followed by the L-DTX group (27.49 ± 7.32), and the Control group (24.52 ± 8.11) having the shortest, with the difference being statistically significant (p < 0.05). (2) Following ED treatment, compared to rats treated with normal saline, the escape latency of the L-DTX (12.00 ± 2.79 vs. 10.77 ± 3.97, p < 0.05), and the H-DTX (12.52 ± 3.69 vs. 9.11 ± 2.88, p < 0.05) rats were significantly shortened. The residence time in the target quadrant of H-DTX rats was significantly prolonged (40.49 ± 5.82 vs. 55.25 ± 6.78, p < 0.05). The CNS damage in the L-DTX rats was repaired to a certain extent during the interval between the two water maze tests (28.89 ± 7.92 vs. 12.00 ± 2.79, p < 0.05). (3) The fractional anisotropy (FA) value of DTI in the hippocampus of rats in the different groups showed variable trends. After treatment with ED, though the FA values of most areas in the hippocampus of rats in L-DTX and H-DTX groups were higher than before, they did not reach the normal level.ConclusionED can ameliorate the cognitive dysfunctions caused by DTX in rats by improving the learning and memory impairment, which is reflected in the recovery of biological behavior and DTI indicators of the hippocampus.
Collapse
Affiliation(s)
- Ping Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Hai Liu
- Department of Urology Surgery, The People’s Hospital of Qijiang District, Chongqing, China
| | - Lijun Wei
- Department of Urology Surgery, The People’s Hospital of Qijiang District, Chongqing, China
| | - Xun Shi
- Department of Nuclear Medicine, The First People’s Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China
| | - Wei Wang
- Department of Nuclear Medicine, The First People’s Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China
| | - Shengxiang Yan
- Department of Science and Technology, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, China
| | - Wenya Zhou
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jiangong Zhang
- Department of Nuclear Medicine, The First People’s Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China
- *Correspondence: Jiangong Zhang, ; Suxia Han,
| | - Suxia Han
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
- *Correspondence: Jiangong Zhang, ; Suxia Han,
| |
Collapse
|
23
|
Gao X, Chen F, Xu X, Liu J, Dong F, Liu Y. Ro25-6981 alleviates neuronal damage and improves cognitive deficits by attenuating oxidative stress via the Nrf2/ARE pathway in ischemia/reperfusion rats. J Stroke Cerebrovasc Dis 2023; 32:106971. [PMID: 36586245 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106971] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Oxidative stress plays a crucial role in the initiation and progression of cerebral ischemia‒reperfusion injury (CIRI). Therefore, ameliorating oxidative damage is considered to be a beneficial strategy for the treatment of CIRI. NMDAR NR2B subunit antagonists have been reported to be beneficial for synaptic plasticity, neuropathic pain, epilepsy, and cerebral ischemia. However, it remains unclear whether the NR2B subunit antagonist Ro25-6981 has any effect on CIRI. METHODS In this study, the Morris water maze test and passive avoidance test were used to detect spatial learning and memory. Neuronal loss was measured by Nissl staining. The expression of NSE was assayed by immunohistochemistry. The activities of MDA, 8-OHdG, SOD, GSH-Px, GST and CAT were detected by assay kits. Real-time PCR was used to detect the mRNA levels of hippocampal SOD, GSH-Px and HO-1. Western blotting was used to measure the activation of the Nrf2/ARE pathway by Ro25-6981. RESULTS Ro25-6981 ameliorated cognitive deficits and neuronal damage induced by ischemia‒reperfusion (I/R). Neuronal injury was decreased and the expression of NSE was increased in the CA1 regions of the hippocampus of I/R rats after Ro25-6981 treatment. Moreover, Ro25-6981 alleviated the levels of MDA and 8-OHdG by elevating the activities of SOD, GSH-Px, GST and CAT. Meanwhile, the mRNA levels of SOD, GSH-Px and HO-1 were increased in I/R rats after Ro25-6981 treatment. Furthermore, Ro25-6981 promoted the translocation of Nrf2 to the nucleus, promoting the expression of the Nrf2 downstream genes HO-1 and NQO1. CONCLUSION The present study indicated that the improvement in the antioxidant properties of Ro25-6981 is mediated by the Nrf2/ARE pathway. This is the first study to demonstrate a favorable effect of Ro25-6981 on cognitive impairment in a CIRI rat model, rendering this NR2B subunit antagonist a promising agent for the treatment or prevention of CIRI.
Collapse
Affiliation(s)
- Xiuxian Gao
- Department of Neurology, The First People's Hospital of Jiujiang, 48 Taling South Road, Jiujiang, Jiangxi Province 332000, China
| | - Fei Chen
- The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221004, China
| | - Xinqi Xu
- The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221004, China
| | - Jinfeng Liu
- School of Life Science, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221004, China
| | - Fuxing Dong
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Yaping Liu
- Laboratory of National Experimental Teaching and Demonstration Center of Basic Medicine, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221004, China.
| |
Collapse
|
24
|
Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil Extracellular Traps in Cerebral Ischemia/Reperfusion Injury: Friend and Foe. Curr Neuropharmacol 2023; 21:2079-2096. [PMID: 36892020 PMCID: PMC10556361 DOI: 10.2174/1570159x21666230308090351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 03/10/2023] Open
Abstract
Cerebral ischemic injury, one of the leading causes of morbidity and mortality worldwide, triggers various central nervous system (CNS) diseases, including acute ischemic stroke (AIS) and chronic ischemia-induced Alzheimer's disease (AD). Currently, targeted therapies are urgently needed to address neurological disorders caused by cerebral ischemia/reperfusion injury (CI/RI), and the emergence of neutrophil extracellular traps (NETs) may be able to relieve the pressure. Neutrophils are precursors to brain injury following ischemic stroke and exert complicated functions. NETs extracellularly release reticular complexes of neutrophils, i.e., double-stranded DNA (dsDNA), histones, and granulins. Paradoxically, NETs play a dual role, friend and foe, under different conditions, for example, physiological circumstances, infection, neurodegeneration, and ischemia/reperfusion. Increasing evidence indicates that NETs exert anti-inflammatory effects by degrading cytokines and chemokines through protease at a relatively stable and moderate level under physiological conditions, while excessive amounts of NETs release (NETosis) irritated by CI/RI exacerbate the inflammatory response and aggravate thrombosis, disrupt the blood-brain barrier (BBB), and initiates sequential neuron injury and tissue damage. This review provides a comprehensive overview of the machinery of NETs formation and the role of an abnormal cascade of NETs in CI/RI, as well as other ischemia-induced neurological diseases. Herein, we highlight the potential of NETs as a therapeutic target against ischemic stroke that may inspire translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Rui Fang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| |
Collapse
|
25
|
Nrf2 Regulates Oxidative Stress and Its Role in Cerebral Ischemic Stroke. Antioxidants (Basel) 2022; 11:antiox11122377. [PMID: 36552584 PMCID: PMC9774301 DOI: 10.3390/antiox11122377] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
Cerebral ischemic stroke is characterized by acute ischemia in a certain part of the brain, which leads to brain cells necrosis, apoptosis, ferroptosis, pyroptosis, etc. At present, there are limited effective clinical treatments for cerebral ischemic stroke, and the recovery of cerebral blood circulation will lead to cerebral ischemia-reperfusion injury (CIRI). Cerebral ischemic stroke involves many pathological processes such as oxidative stress, inflammation, and mitochondrial dysfunction. Nuclear factor erythroid 2-related factor 2 (Nrf2), as one of the most critical antioxidant transcription factors in cells, can coordinate various cytoprotective factors to inhibit oxidative stress. Targeting Nrf2 is considered as a potential strategy to prevent and treat cerebral ischemia injury. During cerebral ischemia, Nrf2 participates in signaling pathways such as Keap1, PI3K/AKT, MAPK, NF-κB, and HO-1, and then alleviates cerebral ischemia injury or CIRI by inhibiting oxidative stress, anti-inflammation, maintaining mitochondrial homeostasis, protecting the blood-brain barrier, and inhibiting ferroptosis. In this review, we have discussed the structure of Nrf2, the mechanisms of Nrf2 in cerebral ischemic stroke, the related research on the treatment of cerebral ischemia through the Nrf2 signaling pathway in recent years, and expounded the important role and future potential of the Nrf2 pathway in cerebral ischemic stroke.
Collapse
|
26
|
Babataheri S, Malekinejad H, Mosarrezaii A, Soraya H. Pre-treatment or post-treatment with hydroxychloroquine demonstrates neuroprotective effects in cerebral ischemia/reperfusion. Fundam Clin Pharmacol 2022; 37:589-598. [PMID: 36433900 DOI: 10.1111/fcp.12856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/21/2022] [Accepted: 11/23/2022] [Indexed: 11/28/2022]
Abstract
Stroke is a serious life-threatening medical condition and is one of the principal reasons for death and disabilities worldwide. The aim of the present study was to determine the neuroprotective effects of hydroxychloroquine (HCQ) and the timing of its administration in cerebral ischemia/reperfusion (I/R) in rats. A global I/R model was used, and HCQ was administered in either pre- or post-treatment doses of 25 and 50 mg/kg. Effects of HCQ on infarct size, histological changes, oxidative stress, and learning and memory were evaluated. Phospho-AMPK and SQSTM1/p62 protein levels were also measured to elucidate the possible mechanisms involved. HCQ in both pre- (at doses of 25 and 50 mg/kg) or post-treatment (at a dose of 50 mg/kg) protocols reduces brain infarct size and histopathological changes and improves learning and memory after cerebral I/R. Pre-treatment with HCQ reduced AMPK activity with no significant effect on SQSTM1/p62 increment. Post-treatment with HCQ increased AMPK activity and SQSTM1/p62 protein levels. Our results show the neuroprotective effects of HCQ on cerebral I/R through the reduction in infarct size, histopathological changes, and improvement in memory and learning functions. Moreover, AMPK and autophagy may play a role in this protective effect.
Collapse
Affiliation(s)
- Shabnam Babataheri
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Hassan Malekinejad
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Arash Mosarrezaii
- Department of Neurology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
27
|
Wang H, Chen X, Miao X, Lu K, He M, Wu X. Dendrobium mixture improves gestational diabetes mellitus through regulating Nrf2/HO1 signaling pathway. Biomed Pharmacother 2022; 155:113656. [PMID: 36116251 DOI: 10.1016/j.biopha.2022.113656] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is characterized by insulin resistance during pregnancy, and it is always combined with serious complications. Dendrobium mixture (DMix) is a kind of traditional Chinese medicine, and it has been proved to be an effective treatment for diabetes. However, the regulatory role of DMix in GDM remains elusive. METHODS High fat feed combined with streptozotocin injection and high glucose medium were used to establish GDM animal and cell models, respectively. The levels of blood glucose, blood lipid, and insulin were measured with commercial kits. Western blotting was used to detect protein expression. RESULTS DMix improved pancreas and placenta injury in GDM rats. DMix reversed the influence of GDM on the levels of SOD, MDA, and glutathione in the serum. Hyperglycemia and hyperlipidemia in GDM rats were suppressed by DMix. The activation of MAPK and inhibition of Nrf2/HO1 in GDM animal and cell models were reversed by DMix. The increase of ROS intensity, apoptosis, and inflammation factors in HG treated cells were reversed by DMix. CONCLUSION This research proved that DMix improved GDM through inhibiting oxidative condition, inflammation factors, hyperglycemia and hyperlipidemia. This study might provide a novel thought for the prevention and treatment of GDM.
Collapse
Affiliation(s)
- Hongri Wang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital to Fujian University of Traditional Chinese Medicine, No. 282, Wusi Road, Fuzhou 350001, Fujian, China.
| | - Xuzheng Chen
- Academy of integratived Medicine, Fujian University of Traditional Chinese Medicine, No. 282, Wusi Road, Fuzhou 350001, Fujian, China.
| | - Xueqin Miao
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital to Fujian University of Traditional Chinese Medicine, No. 282, Wusi Road, Fuzhou 350001, Fujian, China.
| | - Kunbin Lu
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital to Fujian University of Traditional Chinese Medicine, No. 282, Wusi Road, Fuzhou 350001, Fujian, China.
| | - Mengjuan He
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital to Fujian University of Traditional Chinese Medicine, No. 282, Wusi Road, Fuzhou 350001, Fujian, China.
| | - Xiaomei Wu
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital to Fujian University of Traditional Chinese Medicine, No. 282, Wusi Road, Fuzhou 350001, Fujian, China.
| |
Collapse
|
28
|
Edaravone Dexborneol Downregulates Neutrophil Extracellular Trap Expression and Ameliorates Blood-Brain Barrier Permeability in Acute Ischemic Stroke. Mediators Inflamm 2022; 2022:3855698. [PMID: 36032782 PMCID: PMC9410976 DOI: 10.1155/2022/3855698] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022] Open
Abstract
Background Our previous work has shown that inflammatory processes play a detrimental role in the pathophysiology of acute ischemic stroke (AIS). Neutrophil extracellular traps (NETs) have been recognized as a key contributor to the proinflammatory response in AIS and could aggravate blood-brain barrier (BBB) damage. Recently, experimental and clinical researches showed that Edaravone Dexborneol (Eda.B), which is comprised of two active ingredients, Edaravone and (+)-Borneol, was effective in treatment of AIS. However, it is not clear whether the effects of Eda.B against AIS are related to NETs and BBB permeability. Methods Experiment 1 was to detect the effects of Eda.B in AIS patients. Serum samples of volunteers and AIS patients were collected before and 3 days after Edaravone Dexborneol treatment. Markers of NETs and occludin were detected by ELISA kit. Experiment 2 was to explore the effects of Eda.B on experimental stroke mice. Male C57BL/6 mice were subjected to distal middle cerebral artery occlusion (MCAO) and treated with vehicle, Eda.B, or DeoxyribonueleaseI (DNase I). After stroke, the neurobehavioral tests, infarct volume, and cerebral blood flow evaluation were determined. Leakage of Evans blue was to assess the integrity of BBB. Western blot, real-time quantitative polymerase chain reaction (RT-qPCR), and immunofluorescence were used to examine the expression of NETs and tight junction- (TJ-) associated proteins. Results Eda.B significantly improved neurological function and cerebral blood flow but reduced infarct volume after experimental stroke. Eda.B downregulated level of NETs in serum samples of AIS patients and tissue samples of MCAO mouse cortex. Eda.B and DNase I alleviated BBB permeability by upregulating TJ-associated proteins. Conclusion NETs are related to the early stage of AIS. Eda.B exerted neuroprotective effects and ameliorated BBB permeability after AIS.
Collapse
|