1
|
Chen HJ, Chen YF, Chen JF, Qian K, Zhu YY, Fang L, Zhang Y, Yang T, Wang GW, Huang PT. Kuanxiong Aerosol Attenuates Ischemic Stroke Injury via Modulation of the TRPV1 Channel. Chin J Integr Med 2025:10.1007/s11655-024-3669-9. [PMID: 39792345 DOI: 10.1007/s11655-024-3669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVE To evaluate the therapeutic effects of Kuanxiong Aerosol (KXA) on ischemic stroke with reperfusion and elucidate the underlying pharmacological mechanisms. METHODS In vivo pharmacological effects on ischemic stroke with reperfusion was evaluated using the transient middle cerebral artery occlusion (t-MCAO) mice model. To evaluate short-term outcome, 30 mice were randomly divided into vehicle group (n=15) and KXA group (n=15). Mice in KXA and vehicle groups received 69 mg KXA and vehicle for 1 day, respectively. To evaluate long-term outcome, 35 mice were randomly divided into sham group (n=5), vehicle group (n=15), and KXA group (n=15). Mice in KXA and vehicle groups received 69 mg KXA and vehicle for 7 days, respectively. Pathological changes in the brain were observed by 2,3,5-triphenyltetrazolium chloride or Nissl stainings, and behavioral assessments, including the Modified Neurologic Severity Score, Bederson score, rotarod test, and adhesive removal test were conducted. The penetration ability of KXA and KX (KXA without propellants) through the blood-brain barrier was assessed both in vitro using a transwell model and in vivo. Furthermore, in vitro effects of KX (5, 10, and 20 µL/L) on oxygen and glucose deprivation/re-oxygenation (OGD/R)-induced injury, transient receptor potential vanilloid type 1 (TRPV1) modulation, calcium influx, and mitochondrial function were explored through Western blot, CCK-8 assay, JC-1 staining, calcium imaging, adenosine triphosphate (ATP) and antioxidant measurements. RESULTS In in vivo experiments, KXA reduced brain infarct volume and neuron loss in t-MCAO mice. Behavioral assessments showed marked improvement in the neurological deficit of t-MCAO mice with KXA treatment (P<0.05 or P<0.01). Additionally, in vitro findings indicated that KX ameliorated OGD/R-induced injury through TRPV1 channel modulation. KX increased cell viability in OGD/R-treated SH-SY5Y cells and prevented OGD/R-induced calcium overload by downregulating TRPV1 expression and constraining calcium influx through TRPV1 (P<0.05 or P<0.01). Furthermore, KXA maintained the membrane potential and function of mitochondria in OGD/R-treated SH-SY5Y cells. CONCLUSIONS KXA could attenuate ischemic stroke injury through TRPV1 channel modulation, indicating its potential as a promising therapeutic option for stroke in clinical practice.
Collapse
Affiliation(s)
- Hong-Jian Chen
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Ye-Feng Chen
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Ji-Fan Chen
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Kai Qian
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Yang-Yang Zhu
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Lei Fang
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Ying Zhang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tao Yang
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Guo-Wei Wang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Pin-Tong Huang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
2
|
Hou Y, Shang Y, Xu F, Li T, Li M, Wei L, Fan S, Hou W, Gou W, Shang H, Li Y. Ionizing radiation induces neurotoxicity in Xenopus laevis embryos through neuroactive ligand-receptor interaction pathway. ENVIRONMENTAL RESEARCH 2024; 256:119237. [PMID: 38810829 DOI: 10.1016/j.envres.2024.119237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/14/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Ionizing radiation (IR) poses a significant threat to both the natural environment and biological health. Exposure to specific doses of ionizing radiation early in an organism's development can lead to developmental toxicity, particularly neurotoxicity. Through experimentation with Xenopus laevis (X. laevis), we examined the effects of radiation on early developmental stage. Our findings revealed that radiation led to developmental abnormalities and mortality in X. laevis embryos in a dose-dependent manner, disrupting redox homeostasis and inducing cell apoptosis. Additionally, radiation caused neurotoxic effects, resulting in abnormal behavior and neuron damage in the embryos. Further investigation into the underlying mechanisms of radiation-induced neurotoxicity indicated the potential involvement of the neuroactive ligand-receptor interaction pathway, which was supported by RNA-Seq analysis. Validation of gene expression associated with this pathway and analysis of neurotransmitter levels confirmed our hypothesis. In addition, we further validated the important role of this signaling pathway in radiation-induced neurotoxicity through edaravone rescue experiments. This research establishes a valuable model for radiation damage studying and provides some insight into radiation-induced neurotoxicity mechanisms.
Collapse
Affiliation(s)
- Yue Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Yue Shang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Feifei Xu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Tingyang Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Min Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Ling Wei
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Saijun Fan
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Wenbin Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Wenfeng Gou
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China.
| | - Haihua Shang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China.
| | - Yiliang Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China.
| |
Collapse
|
3
|
Zhang Y, Zhang P, Zhang X, Liu Y. HH-A, a modified honokiol, protects against cerebral ischemia/reperfusion induced brain injury in rodent via Nrf2/HO-1 signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3389-3402. [PMID: 37955691 DOI: 10.1007/s00210-023-02816-8] [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: 05/22/2023] [Accepted: 10/22/2023] [Indexed: 11/14/2023]
Abstract
Honokiol, a bioactive component found in Magnolia officinalis, has shown in protecting against ischemic stroke in animal models. However, its poor water solubility has limited its clinical applications. In this study, we introduced a hydrophilic building block on the aromatic ring of honokiol, resulting in the synthesis of four new compounds (HH-A, -B, -C and -D) with significantly improved water solubility. We then investigated the neuroprotective effects of these compounds in mouse and rat models of transient middle cerebral artery occlusion/reperfusion (tMCAO/R) brain injury. Among the compounds tested, HH-A, also known as (S)-6-((3',5-diallyl-2,4'-dihydroxy-[1,1'-biphenyl]-3-yl)amino)-6-oxohexane-1,5-diaminium chloride, showed the most promising results. HH-A was found to significantly reduced the infarct volume and brain edema in mice. It also outperformed the other three compounds and honokiol, even surpassing the effects of edaravone dexborneol. Additionally, HH-A demonstrated dose-dependent improvements in body weight, neurological deficits, and infarct volume. Further analysis in tMCAO/R rat model revealed that HH-A treatment led to significant upregulations of Nrf2 and HO-1 in the brain. HH-A also significantly reduced the expression of HNE, and exhibited anti-apoptotic effects by decreasing the expression of Bax and increasing the expression of Bcl-2. This was further supported by a decrease in the number of TUNEL positive cells. Taken together, the neuroprotective effects of HH-A may be attributed to its ability to target the Nrf2/HO-1 signaling pathway, leading to reduced oxidative stress and apoptosis in the brain. These findings suggest that HH-A has potential as a therapeutic agent for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yuying Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin, 300070, China
- Beijing Honghui Meditech Co., Ltd, No. 50 Huatuo Road, CBP Daxing, Beijing, 102600, China
| | - Pingping Zhang
- Beijing Honghui Meditech Co., Ltd, No. 50 Huatuo Road, CBP Daxing, Beijing, 102600, China
| | - Xin Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin, 300070, China.
| | - Ye Liu
- Beijing Honghui Meditech Co., Ltd, No. 50 Huatuo Road, CBP Daxing, Beijing, 102600, China.
| |
Collapse
|
4
|
Duranti E, Cordani N, Villa C. Edaravone: A Novel Possible Drug for Cancer Treatment? Int J Mol Sci 2024; 25:1633. [PMID: 38338912 PMCID: PMC10855093 DOI: 10.3390/ijms25031633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in understanding the causes and progression of tumors, cancer remains one of the leading causes of death worldwide. In light of advances in cancer therapy, there has been a growing interest in drug repurposing, which involves exploring new uses for medications that are already approved for clinical use. One such medication is edaravone, which is currently used to manage patients with cerebral infarction and amyotrophic lateral sclerosis. Due to its antioxidant and anti-inflammatory properties, edaravone has also been investigated for its potential activities in treating cancer, notably as an anti-proliferative and cytoprotective drug against side effects induced by traditional cancer therapies. This comprehensive review aims to provide updates on the various applications of edaravone in cancer therapy. It explores its potential as a standalone antitumor drug, either used alone or in combination with other medications, as well as its role as an adjuvant to mitigate the side effects of conventional anticancer treatments.
Collapse
Affiliation(s)
| | | | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.D.); (N.C.)
| |
Collapse
|
5
|
Kovács KB, Bencs V, Hudák L, Oláh L, Csiba L. Hemorrhagic Transformation of Ischemic Strokes. Int J Mol Sci 2023; 24:14067. [PMID: 37762370 PMCID: PMC10531605 DOI: 10.3390/ijms241814067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Ischemic stroke, resulting from insufficient blood supply to the brain, is among the leading causes of death and disability worldwide. A potentially severe complication of the disease itself or its treatment aiming to restore optimal blood flow is hemorrhagic transformation (HT) increasing morbidity and mortality. Detailed summaries can be found in the literature on the pathophysiological background of hemorrhagic transformation, the potential clinical risk factors increasing its chance, and the different biomarkers expected to help in its prediction and clinical outcome. Clinicopathological studies also contribute to the improvement in our knowledge of hemorrhagic transformation. We summarized the clinical risk factors of the hemorrhagic transformation of ischemic strokes in terms of risk reduction and collected the most promising biomarkers in the field. Also, auxiliary treatment options in reperfusion therapies have been reviewed and collected. We highlighted that the optimal timing of revascularization treatment for carefully selected patients and the individualized management of underlying diseases and comorbidities are pivotal. Another important conclusion is that a more intense clinical follow-up including serial cranial CTs for selected patients can be recommended, as clinicopathological investigations have shown HT to be much more common than clinically suspected.
Collapse
Affiliation(s)
| | | | | | | | - László Csiba
- Department of Neurology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (K.B.K.); (V.B.); (L.H.); (L.O.)
| |
Collapse
|
6
|
Mangiferin: the miraculous xanthone with diverse pharmacological properties. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:851-863. [PMID: 36656353 DOI: 10.1007/s00210-022-02373-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023]
Abstract
Mangiferin (1,3,6,7-tetrahydroxy-2-[3,4,5-trihydroxy-6-(hydroxymethyl) oxan-2-yl] xanthen-9-one) is a bioactive component derived primarily from the mango tree. Belonging to the Xanthone family, its structure allows it to engage with a variety of pharmacological targets. The symmetric linked core of xanthones has a heterogeneous biogenetic background. The carbon atoms are designated in a biochemical order, which reveals the reason of ring A (C1-C4) being referred to as acetate originated, and ring B (C5-C8) is referred to as shikimate originated. The antibacterial, hypocholesterolemic, antiallergic, cardiotonic, antidiabetic, anti-neoplastic, neuroprotective, antioxidant and immunomodulatory properties have all been demonstrated for the secondary metabolite. This study assessed and explained the important medical properties of mangiferin available in published literature, as well as its natural source, biosynthesis, absorption and bioavailability; multiple administration routes; metabolism; nanotechnology for enhanced efficacy of mangiferin and its toxicity, to aid the anticipated on-going potential of mangiferin as a novel diagnostic treatment.
Collapse
|
7
|
Edaravone Dexborneol Alleviates Cerebral Ischemic Injury via MKP-1-Mediated Inhibition of MAPKs and Activation of Nrf2. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4013707. [PMID: 36110124 PMCID: PMC9470337 DOI: 10.1155/2022/4013707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/27/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
The edaravone and dexborneol concentrated solution for injection (edaravone-dexborneol) is a medication used clinically to treat neurological impairment induced by ischemic stroke. This study was aimed at investigating the preventive effects and the underlying mechanisms of edaravone-dexborneol on cerebral ischemic injury. A rat four-vessel occlusion (4-VO) model was established, and the neuronal injury and consequent neurological impairment of rats was investigated. Brain tissue malondialdehyde (MDA), myeloperoxidase (MPO), and nitric oxide (NO) levels were determined. The levels of proteins in mitogen-activated protein kinases (MAPKs), nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor-κB (NF-κB) signaling pathways were determined by western immunoblotting. The function of mitogen-activated protein kinase phosphatase 1 (MKP-1) was investigated using both western blot and immunofluorescence methods, and the effect of the MKP-1 inhibitor, (2E)-2-benzylidene-3-(cyclohexylamino)-3H-inden-1-one (BCI), was investigated. The results indicated that edaravone-dexborneol alleviated neurological deficiency symptoms and decreased apoptosis and neuron damage in the hippocampal CA1 area of the ischemic rats. Edaravone-dexborneol increased the MKP-1 level; decreased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK); inhibited NF-κB p65 activation; and boosted Nrf2 activation, all of which were partially reversed by the MKP-1 inhibitor, BCI. The above results indicated that the upregulation of MKP-1 contributed to the protective effects of edaravone-dexborneol against ischemic brain injury. Our findings support the hypothesis that edaravone-dexborneol can alleviate cerebral ischemic injury via the upregulation of MKP-1, which inhibits MAPKs and activates Nrf2.
Collapse
|
8
|
Liu W, Wang L, Liu C, Dai Z, Li T, Tang B. Edaravone Ameliorates Cerebral Ischemia–Reperfusion Injury by Downregulating Ferroptosis <i>via</i> the Nrf2/FPN Pathway in Rats. Biol Pharm Bull 2022; 45:1269-1275. [DOI: 10.1248/bpb.b22-00186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Wenpeng Liu
- Medical School, Hunan University of Chinese Medicine
| | - Linlin Wang
- Medical School, Hunan University of Chinese Medicine
| | - Canwen Liu
- Medical School, Hunan University of Chinese Medicine
| | - Ziwei Dai
- Medical School, Hunan University of Chinese Medicine
| | - Tenglong Li
- The Second Affiliated Hospital of Hunan University of Chinese Medicine
| | - Biao Tang
- Medical School, Hunan University of Chinese Medicine
| |
Collapse
|
9
|
Alterations of inflammatory cytokines in super-acute stroke patients and the potential pathogenesis. J Clin Neurosci 2022; 99:35-43. [DOI: 10.1016/j.jocn.2022.02.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 11/19/2022]
|
10
|
Singh S, Gautam U, Manvi FV. Protective Impact of Edaravone Against ZnO NPs-induced Oxidative Stress in the Human Neuroblastoma SH-SY5Y Cell Line. Cell Mol Neurobiol 2022; 42:1189-1210. [PMID: 33222098 PMCID: PMC11441218 DOI: 10.1007/s10571-020-01011-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022]
Abstract
Extensive applications of ZnO NPs (zinc oxide nanoparticles) in daily life have created concern about their biotoxicity. Zinc oxide nanoparticles induce oxidative stress, inflammation, and apoptosis in neurons. Edaravone applies antioxidant agent and anti-inflammatory impacts in the different cells, as evaluated in both in vitro and in vivo experimental models. This study is designed to explore, how edaravone would avert mitochondrial impairment in human neuronal cells against ZnO NPs-induced toxicity. Accordingly, we analyzed here whether a pretreatment (for 24 h) with edaravone (10-100 μM) would enhance mitochondrial protection in the human neuroblastoma cells SH-SY5Y against ZnO NPs-induced toxicity. We found that edaravone at 25 μM averted the ZnO NPs-induced decrease in the amounts of adenosine triphosphate (ATP), just as on the activity of the complexes I and V. Also, edaravone induced an antioxidant activity by diminishing the levels of lipid peroxidation, protein carbonylation, and protein nitration in the mitochondrial membranes. Edaravone blocked the ZnO NPs-induced transcription factor nuclear factor-κB (NF-κB) upregulation. The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 μM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. After this examination, it can be concluded that edaravone caused cytoprotective impacts in an HO-1-dependent manner in SH-SY5Y cells against ZnO NPs-induced toxicity.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Hajipur, Bihar, India.
- K.L.E. Academy of Higher Education & Research, Belagavi, Karnataka, India.
| | - Upendr Gautam
- Vinayaka Mission's Research Foundation, Ariyanur, Tamil Nadu, India
| | - F V Manvi
- K.L.E. Academy of Higher Education & Research, Belagavi, Karnataka, India
| |
Collapse
|
11
|
Xu H, Shen J, Xiao J, Chen F, Wang M. Neuroprotective effect of cajaninstilbene acid against cerebral ischemia and reperfusion damages by activating AMPK/Nrf2 pathway. J Adv Res 2021; 34:199-210. [PMID: 35024191 PMCID: PMC8655138 DOI: 10.1016/j.jare.2020.07.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Ischemic stroke is one of the leading causes of death worldwide. Recently, neuroprotection is regarded as an important preventative and therapeutic strategy for ischemic stroke. Cajaninstilbene acid (CSA), a unique stilbenoid with a styryl group, is a potential neuroprotective agent. OBJECTIVES Hence, this study aimed to evaluate the neuroprotective effect and molecular mechanism of CSA against cerebral ischemia/reperfusion (I/R) damages. METHODS Cerebral ischemia was modeled by oxygen and glucose deprivation (OGD) in SH-SY5Y cells or transient intraluminal suture middle cerebral artery occlusion (MCAO) in rats, and tert-butyl hydroperoxide (t-BHP) was used to induce oxidative stress in SH-SY5Y cells. CSA (2.5, 5 mg/kg) was intraperitoneally given upon reperfusion after 2 h of MCAO. The signaling pathways were analyzed by Western blotting and inhibitor blocking. RESULTS CSA possessed significant neuroprotective activity, as evidenced by the reduced cell death in OGD/R or t-BHP injured SH-SY5Y cells, and decreased infarct volume and neurological deficits in MCAO/R rats. Further studies indicated that the protective effect was achieved via the antioxidant activity of CSA, which decreased the oxidative stress and its related mitochondrial dysfunction in SH-SY5Y cells. Notably, Nrf2 was activated in SH-SY5Y cells and MCAO/R rats by CSA, and the inhibition of Nrf2 by brusatol weakened CSA-mediated neuroprotection. Furthermore, after applying a series of kinase inhibitors, CSA-induced Nrf2 activation was markedly inhibited by BML-275 (an AMPK inhibitor), implying that AMPK was the dominant kinase to regulate the Nrf2 pathway for CSA's neuroprotective effects with enhanced AMPK phosphorylation observed both in vivo and in vitro. CONCLUSION CSA exerted neuroprotection via activating the AMPK/Nrf2 pathway to reduce I/R-induced cellular oxidative stress and mitochondrial disfunction. CSA could be a potential neuroprotective drug candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hui Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Mingfu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
12
|
Shakkour Z, Issa H, Ismail H, Ashekyan O, Habashy KJ, Nasrallah L, Jourdi H, Hamade E, Mondello S, Sabra M, Zibara K, Kobeissy F. Drug Repurposing: Promises of Edaravone Target Drug in Traumatic Brain Injury. Curr Med Chem 2021; 28:2369-2391. [PMID: 32787753 DOI: 10.2174/0929867327666200812221022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 11/22/2022]
Abstract
Edaravone is a potent free-radical scavenger that has been in the market for more than 30 years. It was originally developed in Japan to treat strokes and has been used there since 2001. Aside from its anti-oxidative effects, edaravone demonstrated beneficial effects on proinflammatory responses, nitric oxide production, and apoptotic cell death. Interestingly, edaravone has shown neuroprotective effects in several animal models of diseases other than stroke. In particular, edaravone administration was found to be effective in halting amyotrophic lateral sclerosis (ALS) progression during the early stages. Accordingly, after its success in Phase III clinical studies, edaravone has been approved by the FDA as a treatment for ALS patients. Considering its promises in neurological disorders and its safety in patients, edaravone is a drug of interest that can be repurposed for traumatic brain injury (TBI) treatment. Drug repurposing is a novel approach in drug development that identifies drugs for purposes other than their original indication. This review presents the biochemical properties of edaravone along with its effects on several neurological disorders in the hope that it can be adopted for treating TBI patients.
Collapse
Affiliation(s)
- Zaynab Shakkour
- American University of Beirut, Faculty of Medicine, Department of Biochemistry and Molecular Genetics, Beirut, Lebanon
| | - Hawraa Issa
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Helene Ismail
- American University of Beirut, Faculty of Medicine, Department of Biochemistry and Molecular Genetics, Beirut, Lebanon
| | - Ohanes Ashekyan
- American University of Beirut, Faculty of Medicine, Department of Biochemistry and Molecular Genetics, Beirut, Lebanon
| | - Karl John Habashy
- Faculty of Medicine, American, University of Beirut, Beirut, Lebanon
| | - Leila Nasrallah
- American University of Beirut, Faculty of Medicine, Department of Biochemistry and Molecular Genetics, Beirut, Lebanon
| | - Hussam Jourdi
- Biology & Environmental Sciences Division at University of Balamand, Souk El Gharb, Aley, Lebanon
| | - Eva Hamade
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mirna Sabra
- Faculty of Medicine, Lebanese University, Neuroscience Research Center (NRC), Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Firas Kobeissy
- American University of Beirut, Faculty of Medicine, Department of Biochemistry and Molecular Genetics, Beirut, Lebanon
| |
Collapse
|
13
|
Yakypova LR, Baeva LA, Safiullin RL. Reactivity of Peroxyl Radicals with 5-Methyl-4-[(Propylsulfanyl)methyl]-2,4-Dihydro-3H-Pyrazol-3-one. KINETICS AND CATALYSIS 2021. [DOI: 10.1134/s0023158421010134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
14
|
Lee TK, Shin MC, Ahn JH, Kim DW, Kim B, Sim H, Lee JC, Cho JH, Park JH, Kim YM, Won MH, Lee CH. CD200 Change Is Involved in Neuronal Death in Gerbil Hippocampal CA1 Field Following Transient Forebrain Ischemia and Postischemic Treatment with Risperidone Displays Neuroprotection without CD200 Change. Int J Mol Sci 2021; 22:ijms22031116. [PMID: 33498705 PMCID: PMC7865463 DOI: 10.3390/ijms22031116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 11/16/2022] Open
Abstract
It has been reported that CD200 (Cluster of Differentiation 200), expressed in neurons, regulates microglial activation in the central nervous system, and a decrease in CD200 expression causes an increase in microglial activation and neuronal loss. The aim of this study was to investigate time-dependent changes in CD200 expression in the hippocampus proper (CA1, 2, and 3 fields) after transient forebrain ischemia for 5 min in gerbils. In this study, 5-min ischemia evoked neuronal death (loss) of pyramidal neurons in the CA1 field, but not in the CA2/3 fields, at 5 days postischemia. In the sham group, CD200 expression was found in pyramidal neurons of the CA1 field, and the immunoreactivity in the group with ischemia was decreased at 6 h postischemia, dramatically increased at 12 h postischemia, decreased (to level found at 6 h postischemia) at 1 and 2 days postischemia, and significantly increased again at 5 days postischemia. At 5 days postischemia, CD200 immunoreactivity was strongly expressed in microglia and GABAergic neurons. However, in the CA3 field, the change in CD200 immunoreactivity in pyramidal neurons was markedly weaker than that in the CA1 field, showing there was no expression of CD 200 in microglia and GABAergic neurons. In addition, treatment of 10 mg/kg risperidone (an atypical antipsychotic drug) after the ischemia hardly changed CD200 immunoreactivity in the CA1 field, showing that CA1 pyramidal neurons were protected from the ischemic injury. These results indicate that the transient ischemia-induced change in CD200 expression may be associated with specific and selective neuronal death in the hippocampal CA1 field following transient forebrain ischemia.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Biomedical Science and Research, Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Gangwon, Korea;
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Gangwon, Korea; (M.C.S.); (J.H.C.)
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Gyeongnam, Korea;
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung 25457, Gangwon, Korea;
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Jun Hwi Cho
- Department of Emergency Medicine, Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Gangwon, Korea; (M.C.S.); (J.H.C.)
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Gyeongbuk, Korea;
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
- Correspondence: (M.-H.W.); (C.-H.L.); Tel.: +82-33-250-8891 (M.-H.W.); +82-41-550-1441 (C.-H.L.); Fax: +82-33-256-1614 (M.-H.W.); +82-41-559-7899 (C.-H.L.)
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Chungnam, Korea
- Correspondence: (M.-H.W.); (C.-H.L.); Tel.: +82-33-250-8891 (M.-H.W.); +82-41-550-1441 (C.-H.L.); Fax: +82-33-256-1614 (M.-H.W.); +82-41-559-7899 (C.-H.L.)
| |
Collapse
|
15
|
Ma G, Pan Z, Kong L, Du G. Neuroinflammation in hemorrhagic transformation after tissue plasminogen activator thrombolysis: Potential mechanisms, targets, therapeutic drugs and biomarkers. Int Immunopharmacol 2020; 90:107216. [PMID: 33296780 DOI: 10.1016/j.intimp.2020.107216] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/18/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022]
Abstract
Hemorrhagic transformation (HT) is a common and serious complication following ischemic stroke, especially after tissue plasminogen activator (t-PA) thrombolysis, which is associated with increased mortality and disability. Due to the unknown mechanisms and targets of HT, there are no effective therapeutic drugs to decrease the incidence of HT. In recent years, many studies have found that neuroinflammation is closely related to the occurrence and development of HT after t-PA thrombolysis, including glial cell activation in the brain, peripheral inflammatory cell infiltration and the release of inflammatory factors, involving inflammation-related targets such as NF-κB, MAPK, HMGB1, TLR4 and NLRP3. Some drugs with anti-inflammatory activity have been shown to protect the BBB and reduce the risk of HT in preclinical experiments and clinical trials, including minocycline, fingolimod, tacrolimus, statins and some natural products. In addition, the changes in MMP-9, VAP-1, NLR, sICAM-1 and other inflammatory factors are closely related to the occurrence of HT, which may be potential biomarkers for the diagnosis and prognosis of HT. In this review, we summarize the potential inflammation-related mechanisms, targets, therapeutic drugs, and biomarkers associated with HT after t-PA thrombolysis and discuss the relationship between neuroinflammation and HT, which provides a reference for research on the mechanisms, prevention and treatment drugs, diagnosis and prognosis of HT.
Collapse
Affiliation(s)
- Guodong Ma
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zirong Pan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Linglei Kong
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Guanhua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
16
|
Wang B, Lin W. Edaravone Protects against Pancreatic and Intestinal Injury after Acute Pancreatitis via Nuclear Factor-κB Signaling in Mice. Biol Pharm Bull 2020; 43:509-515. [PMID: 32115510 DOI: 10.1248/bpb.b19-00944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acute pancreatitis (AP) is one kind of acute surgical abdominal disease in the world. It causes intestinal damage with subsequent bacterial migration, endotoxemia and secondary pancreatic infections. In this investigation, we determined that edaravone (EDA) reduces pancreatic and intestinal injury after AP in mice. This was demonstrated by a reduction in histological score, apoptosis, interleukin (IL)-6, IL-1β and tumor necrosis factor (TNF)-α, along with obstructing activation of Toll-like receptor 4 (TLR4) and nuclear factor-κB (NFκB). Our study results suggested that EDA exerts its protective effects against pancreatic and intestinal injury after AP via regulation of the TLR4/NFκB pathway. Our findings provide the basis for EDA to treat AP-induced pancreatic and intestinal injury, even might develop as a potential therapy for other inflammatory diseases.
Collapse
Affiliation(s)
- Bingbin Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University
| | - Wendong Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University
| |
Collapse
|
17
|
Yokoyama H, Tsujii M, Iino T, Nakamura T, Sudo A. Inhibitory effect of edaravone on systemic inflammation and local damage in skeletal muscles following long-term ischemia to murine hind limb. J Orthop Surg (Hong Kong) 2020; 27:2309499019874470. [PMID: 31549573 DOI: 10.1177/2309499019874470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PURPOSE The purpose of this study was to evaluate local and systemic pathology in a murine model of ischemia-reperfusion (I/R) injury induced by long-term application of a tourniquet on the hind limbs and to assess the protective effects of edaravone, a potent systemic scavenger of free radicals, using this model. METHODS Sixty C57BL6 mice were divided in two groups, with one group receiving a 3 mg/kg intraperitoneal injection of edaravone and the other group receiving an identical amount of saline 30 min before ischemia under deep anesthesia. The left thigh of each animal was constricted for 4 h with a 4.5-oz. orthodontic rubber band to induce ischemia; 4 h was the critical duration for skeletal muscles. After ischemia, specimens of skeletal muscles, both kidneys, and plasma were collected at 0, 2, 12, 24, 48, and 72 h. Injury to the skeletal muscles and vacuolar degeneration of the kidneys were histologically assessed. Additionally, apoptosis of skeletal muscle cells was assessed by analysis of caspase 3/7 activity and TUNEL staining. Plasma tumor necrosis factor (TNF)-α levels were measured using an enzyme-linked immunosorbent assay kit. RESULTS Skeletal muscles exhibited prominent injury of myofibers at 12 h after I/R injury, with clear upregulation of plasma TNF-α expression and histologic evidence of tubular dysfunction of the kidneys. Plasma TNF-α levels declined and histologic renal damage was ameliorated in edaravone-treated mice, but treatment did not protect skeletal muscle following ischemia for 4 h. Nonetheless, compared with group S, expression of the apoptosis marker caspase 3/7 was significantly inhibited in the skeletal hind limb muscles of Ed-group mice affected by reperfusion injury following ischemia for 4 h. CONCLUSION The present study demonstrated that edaravone is a potentially useful drug for systemic or local treatment of reperfusion injury resulting from long-term ischemia.
Collapse
Affiliation(s)
- Hirokazu Yokoyama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Masaya Tsujii
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiro Iino
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Tomoki Nakamura
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| |
Collapse
|
18
|
Wu L, Xiong X, Wu X, Ye Y, Jian Z, Zhi Z, Gu L. Targeting Oxidative Stress and Inflammation to Prevent Ischemia-Reperfusion Injury. Front Mol Neurosci 2020; 13:28. [PMID: 32194375 PMCID: PMC7066113 DOI: 10.3389/fnmol.2020.00028] [Citation(s) in RCA: 307] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
The cerebral ischemia injury can result in neuronal death and/or functional impairment, which leads to further damage and dysfunction after recovery of blood supply. Cerebral ischemia/reperfusion injury (CIRI) often causes irreversible brain damage and neuronal injury and death, which involves many complex pathological processes including oxidative stress, amino acid toxicity, the release of endogenous substances, inflammation and apoptosis. Oxidative stress and inflammation are interactive and play critical roles in ischemia/reperfusion injury in the brain. Oxidative stress is important in the pathological process of ischemic stroke and is critical for the cascade development of ischemic injury. Oxidative stress is caused by reactive oxygen species (ROS) during cerebral ischemia and is more likely to lead to cell death and ultimately brain death after reperfusion. During reperfusion especially, superoxide anion free radicals, hydroxyl free radicals, and nitric oxide (NO) are produced, which can cause lipid peroxidation, inflammation and cell apoptosis. Inflammation alters the balance between pro-inflammatory and anti-inflammatory factors in cerebral ischemic injury. Inflammatory factors can therefore stimulate or exacerbate inflammation and aggravate ischemic injury. Neuroprotective therapies for various stages of the cerebral ischemia cascade response have received widespread attention. At present, neuroprotective drugs mainly include free radical scavengers, anti-inflammatory agents, and anti-apoptotic agents. However, the molecular mechanisms of the interaction between oxidative stress and inflammation, and their interplay with different types of programmed cell death in ischemia/reperfusion injury are unclear. The development of a suitable method for combination therapy has become a hot topic.
Collapse
Affiliation(s)
- Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Wu
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zeng Zhi
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Demir F, Demir M, Aygun H. Evaluation of the protective effect of edaravone on doxorubicin nephrotoxicity by [ 99mTc]DMSA renal scintigraphy and biochemical methods. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1383-1390. [PMID: 32036411 DOI: 10.1007/s00210-020-01832-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/23/2020] [Indexed: 12/14/2022]
Abstract
To evaluate the nephroprotective effect of edaravone on doxorubicin-induced nephrotoxicity. In this experimental study, twenty-eight Wistar male rats were used. The rats were separated into 4 groups (n = 7); group І (control), rats were treated with saline (4 ml/kg) and group ІІ (doxorubicin), nephrotoxicity was induced by three doses of 18 mg/kg/i.p. doxorubicin, at a 24-h interval on the 12th, 13th, and 14th days. Group ІІІ (edaravone), rats were treated with edaravone (30 mg/kg/for 14 days), and group ІV (edaravone + doxorubicin), rats were treated with edaravone (30 mg/kg/for 14 days) and doxorubicin were injected (18 mg/kg/for 3 days; at a 24-h interval on the 12th, 13th, and 14th days). On the 15th day of the experiment, technetium-99m-labeled dimercaptosuccinic acid ([99mTc]DMSA) uptake was obtained in both kidneys and biochemical parameters from serum and kidney tissue were measured. Doxorubicin led to nephrotoxicity through elevation of serum blood urea nitrogen (BUN), creatinine and tumor necrosis factor-α (TNF-α), nitric oxide (NO), and interleukin-6 (IL-6) in kidney tissue and decreased [99mTc]DMSA uptake level in the kidney when compared with control group (p < 0.01). Pretreatment edaravone significantly decreased BUN and creatinine, also kidney tissue TNF-α, IL-6, NO, and increased [99mTc]DMSA uptake level compared with the doxorubicin. Edaravone has a significant nephroprotective effect through the attenuation of oxidative stress and inflammatory markers during doxorubicin-induced nephrotoxicity in rats.
Collapse
Affiliation(s)
- Fadime Demir
- Department of Nuclear Medicine, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Turkey
| | - Mustafa Demir
- Department of Nephrology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Hatice Aygun
- Department of Physiology, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Turkey.
| |
Collapse
|
20
|
Nakanishi T, Tsujii M, Asano T, Iino T, Sudo A. Protective Effect of Edaravone Against Oxidative Stress in C2C12 Myoblast and Impairment of Skeletal Muscle Regeneration Exposed to Ischemic Injury in Ob/ob Mice. Front Physiol 2020; 10:1596. [PMID: 32009986 PMCID: PMC6974450 DOI: 10.3389/fphys.2019.01596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/19/2019] [Indexed: 12/25/2022] Open
Abstract
Background The aims of this study were to analyze the effects of the administration of edaravone on C2C12 myoblasts exposed to oxidative stress; to evaluate the skeletal muscles in ob/ob mice; and to analyze the effect of the administration of edaravone in the regeneration of skeletal muscle after ischemic injury. Methods In C2C12 myoblasts, oxidative stress was induced by the exposure to 250 μM H2O2 for 4 h with or without pretreatment of 100 μM edaravone. Thereafter, the viability and expression of TNF-α were analyzed by MTS assay and PCR, respectively. Furthermore, an in vivo study was performed on male C57/BL6-ob/ob mice (10 weeks old) and the respective control mice. The skeletal muscles of tibialis anterior and gastrocnemius were excised for histological analysis and TBARS assay after the measurement of blood flow. In addition, the regeneration of the skeletal muscles was analyzed for the expression of MyoD 7 days after the ligation of the right femoral artery. Results Edaravone significantly inhibited the reduction of the viability as well as upregulation of TNF-α expression by treatment with H2O2. In ob/ob mice, wet weight of muscles was significantly lower than that in control mice. In histology, ob/ob mice had significantly less multi-angle shaped myofibers and a significantly high level of MDA. Furthermore, MyoD expression was lower in ob/ob mice than in control mice after the ischemic injury, while edaravone (3 mg/kg) increasingly enhanced MyoD expression. Conclusion Edaravone attenuated the oxidative stress on C2C12 myoblasts, and was effective to regeneration of skeletal muscles after ischemia in ob/ob mice.
Collapse
Affiliation(s)
- Takuya Nakanishi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Masaya Tsujii
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiro Asano
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiro Iino
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| |
Collapse
|
21
|
Dhir N, Medhi B, Prakash A, Goyal MK, Modi M, Mohindra S. Pre-clinical to Clinical Translational Failures and Current Status of Clinical Trials in Stroke Therapy: A Brief Review. Curr Neuropharmacol 2020; 18:596-612. [PMID: 31934841 PMCID: PMC7457423 DOI: 10.2174/1570159x18666200114160844] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/31/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022] Open
Abstract
In stroke (cerebral ischemia), despite continuous efforts both at the experimental and clinical level, the only approved pharmacological treatment has been restricted to tissue plasminogen activator (tPA). Stroke is the leading cause of functional disability and mortality throughout worldwide. Its pathophysiology starts with energy pump failure, followed by complex signaling cascade that ultimately ends in neuronal cell death. Ischemic cascade involves excessive glutamate release followed by raised intracellular sodium and calcium influx along with free radicals' generation, activation of inflammatory cytokines, NO synthases, lipases, endonucleases and other apoptotic pathways leading to cell edema and death. At the pre-clinical stage, several agents have been tried and proven as an effective neuroprotectant in animal models of ischemia. However, these agents failed to show convincing results in terms of efficacy and safety when the trials were conducted in humans following stroke. This article highlights the various agents which have been tried in the past but failed to translate into stroke therapy along with key points that are responsible for the lagging of experimental success to translational failure in stroke treatment.
Collapse
Affiliation(s)
| | - Bikash Medhi
- Address correspondence to this author at the Department of Pharmacology, Research Block B, 4th Floor, Room no 4043, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India; E-mail:
| | | | | | | | | |
Collapse
|
22
|
Yamato SH, Nakamura S, Htun Y, Nakamura M, Jinnai W, Nakao Y, Mitsuie T, Koyano K, Wakabayashi T, Morimoto AH, Sugino M, Iwase T, Kondo SI, Yasuda S, Ueno M, Miki T, Kusaka T. Intravenous Edaravone plus Therapeutic Hypothermia Offers Limited Neuroprotection in the Hypoxic-Ischaemic Newborn Piglet. Neonatology 2020; 117:713-720. [PMID: 33113527 DOI: 10.1159/000511085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/22/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Therapeutic hypothermia (TH) is a standard therapy for neonatal hypoxic-ischaemic encephalopathy. One potential additional therapy is the free radical scavenger edaravone (EV; 3-methyl-1-phenyl-2-pyrazolin-5-one). OBJECTIVES AND METHODS This study aimed to compare the neuroprotective effects of edaravone plus therapeutic hypothermia (TH + EV) with those of TH alone after a hypoxic-ischaemic insult in the newborn piglet. Anaesthetized piglets were subjected to 40 min of hypoxia (3-5% inspired oxygen), and cerebral ischaemia was assessed using cerebral blood volume. Body temperature was maintained at 39.0 ± 0.5°C in the normothermia group (NT, n = 8) and at 33.5 ± 0.5°C (24 h after the insult) in the TH (n = 7) and TH + EV (3 mg/kg intravenous every 12 h for 3 days after the insult; n = 6) groups under mechanical ventilation. RESULTS Five days after the insult, the mean (standard deviation) neurological scores were 10.9 (5.7) in the NT group, 17.0 (0.4) in the TH group (p = 0.025 vs. NT), and 15.0 (3.9) in the TH + EV group. The histopathological score of the TH + EV group showed no significant improvement compared with that of the other groups. CONCLUSION TH + EV had no additive neuroprotective effects after hypoxia-ischaemia in neurological and histopathological assessments.
Collapse
Affiliation(s)
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan,
| | - Yinmon Htun
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Makoto Nakamura
- Department of Neonatology, National Hospital Organization Okayama Medical Center, Okayam, Japan
| | - Wataru Jinnai
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yasuhiro Nakao
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tsutomu Mitsuie
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | | | | - Masashiro Sugino
- Division of Neonatology, Shikoku Medical Center for Children and Adults, Kagawa, Japan
| | - Takashi Iwase
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Sonoko Ijichi Kondo
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Saneyuki Yasuda
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
23
|
Higashi H, Kinjo T, Uno K, Kuramoto N. Regulatory effects associated with changes in intracellular potassium level in susceptibility to mitochondrial depolarization and excitotoxicity. Neurochem Int 2019; 133:104627. [PMID: 31805298 DOI: 10.1016/j.neuint.2019.104627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/16/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
Excitotoxicity has been believed to be one of the causes of neurodegenerative diseases such as Alzheimer's disease and Huntington's disease. So far, much research has been done to suppress the neuronal excessive excitations, however, we still have not achieved full control, which may be due to the lack of some factors. As a matter of course, there is an urgent need to clarify all mechanisms that inhibit the onset and progression of neurodegenerative diseases. We found that potassium ion level regulation may be important in the sense that it suppresses mitochondrial depolarization rather than hyperpolarization of cell membrane potential. Minoxidil, an opener of ATP-activated potassium (KATP) channels decreased injury with middle cerebral artery occlusion in vivo experiment using TTC staining. In the primary cortical neurons, N-methyl-D-aspartate (NMDA)-induced mitochondrial depolarization was suppressed by minoxidil treatment. Minoxidil inhibited the increase in levels of cleaved caspase 3 and the release of cytochrome c into the cytosol, further reducing potassium ion levels. It was observed decreased potassium levels in neurons by the treatment of minoxidil. Those effects of minoxidil were blocked by glibenclamide. Therefore, it was suggested that minoxidil, via opening of KATP channels, reduced intracellular potassium ion level that contribute to mitochondrial depolarization, and suppressed subsequent NMDA-induced mitochondrial depolarization. Our findings suggest that the control of ion levels in neurons could dominate the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hiroshi Higashi
- Laboratory of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, 573-0101, Japan
| | - Toshihiko Kinjo
- Laboratory of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, 573-0101, Japan
| | - Kyosuke Uno
- Laboratory of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, 573-0101, Japan
| | - Nobuyuki Kuramoto
- Laboratory of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, 573-0101, Japan.
| |
Collapse
|
24
|
Potential use of edaravone to reduce specific side effects of chemo-, radio- and immuno-therapy of cancers. Int Immunopharmacol 2019; 77:105967. [DOI: 10.1016/j.intimp.2019.105967] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
|
25
|
Miao H, Jiang Y, Geng J, Zhang B, Zhu G, Tang J. Edaravone Administration Confers Neuroprotection after Experimental Intracerebral Hemorrhage in Rats via NLRP3 Suppression. J Stroke Cerebrovasc Dis 2019; 29:104468. [PMID: 31694784 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104468] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/15/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Intracerebral hemorrhage (ICH) is one of the leading causes of disability and mortality in adult, which lacks effective therapies. Edaravone has showed its neuroprotective effects after ischemia stroke, but its effects and possible mechanisms after ICH are poorly understood. Here, we investigated whether edaravone confers neuroprotection after ICH in rats and explored the potential mechanisms involved. METHODS ICH was induced in the right basal ganglia of Sprague-Dawley rats by stereotacticly injection of 200 μl autologous blood. Edaravone (3 mg/kg) or vehicle (saline) was administered intravenously and NLRP3 selective antagonist (MCC950, 10 mg/kg) was intraperitoneally injected to study the potential mechanism. Water Morris Maze Test and Rotarod test were used to elucidate neurological function and Fluoro-Jade C was used to study neurodegeneration after ICH. Western blot assay, Reverse Transcription-Polymerase Chain Reaction (RT-PCR) and immunohistochemistry were used to check the expression of molecules involved. RESULTS As a result, we found that edaravone significantly alleviated brain edema and conferred the neurological deficits of rats after ICH. Hematoma increased NLRP3 expression in microglia, which was decreased by edaravone. Moreover, we demonstrated that edaravone shared a similar effect with MCC950 on alleviating neurodegeneration and decreasing the expression of IL-1β, Caspase 1 and NF-κB in protein or mRNA. Lastly, edaravone and MCC950 both increased the number of Tuj-1 positive neuronal cells peripheral hematoma. CONCLUSIONS The present study demonstrated that edaravone conducted neuroprotection after ICH partially via suppressing NF-κB-dependent NLRP3 in microglia, which contributed a novel evidence for clinic usage of edaravone after ICH.
Collapse
Affiliation(s)
- Hongping Miao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongxiang Jiang
- Department of Neurosurgery, The second affiliated hospital of Chongqing Medical University, Chongqing, China
| | - Junjun Geng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
26
|
A novel free radical scavenger, NSP-116, ameliorated the brain injury in both ischemic and hemorrhagic stroke models. J Pharmacol Sci 2019; 141:119-126. [PMID: 31679961 DOI: 10.1016/j.jphs.2019.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/13/2019] [Accepted: 09/24/2019] [Indexed: 12/28/2022] Open
Abstract
Reperfusion injury is a serious problem in ischemic stroke therapy, which leads to neuronal damage and intracranial hemorrhage (ICH). A novel free radical scavenger, NSP-116, has anti-oxidative effect and may ameliorate reperfusion injury. The purpose of this study was to investigate the effects of NSP-116 on both ischemic and hemorrhagic stroke models. First, we assessed whether NSP-116 has protective effects in vitro. Pre-treatment of NSP-116 decreased neuronal cell damage induced by H2O2 or LPS. Moreover, NSP-116 also suppressed mitochondria damage and apoptosis in H2O2-induced neuronal injury model. Based on these results, we used a middle cerebral artery occlusion (MCAO)-induced ischemic stroke model or a collagenase-induced ICH model. Using the MCAO model, we evaluated the cerebral blood flow (CBF), neurological deficit, and infarct volume. Hematoma volume was assessed at 3 days after ICH. In the MCAO model, oral administration of NSP-116 at 30 mg/kg attenuated the reduction of CBF, neurological deficits, and infarct formation. Interestingly, NSP-116 also ameliorated hematoma expansion and neurological deficits in the ICH model. Additionally, pre-treatment of NSP-116 suppressed the brain microvascular endothelial cell death induced by collagenase treatment. Collectively, our findings indicated that oral administration of NSP-116 attenuates both ischemic and hemorrhagic brain injuries after stroke.
Collapse
|
27
|
Ahn JH, Kim DW, Park JH, Lee TK, Lee HA, Won MH, Lee CH. Expression changes of CX3CL1 and CX3CR1 proteins in the hippocampal CA1 field of the gerbil following transient global cerebral ischemia. Int J Mol Med 2019; 44:939-948. [PMID: 31524247 PMCID: PMC6658004 DOI: 10.3892/ijmm.2019.4273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/08/2019] [Indexed: 01/27/2023] Open
Abstract
Chemokine C-X3-C motif ligand 1 (CX3CL1) and its sole receptor, CX3CR1, are known to be involved in neuronal damage/death following brain ischemia. In the present study, time-dependent expression changes of CX3CL1 and CX3CR1 proteins were investigated in the hippocampal CA1 field following 5 min of transient global cerebral ischemia (tgCI) in gerbils. To induce tgCI in gerbils, bilateral common carotid arteries were occluded for 5 min using aneurysm clips. Expression changes of CX3CL1 and CX3CR1 proteins were assessed at 1, 2 and 5 days after tgCI using western blotting and immunohistochemistry. CX3CL1 immunoreactivity was strong in the CA1 pyramidal cells of animals in the sham operation group. Weak CX3CL1 immunoreactivity was detected at 6 h after tgCI, recovered at 1 day after tgCI and disappeared from 5 days after tgCI. CX3CR1 immunoreactivity was very weak in CA1 pyramidal cells of the sham animals. CX3CR1 immunoreactivity in CA1 pyramidal cells was significantly increased at 1 days after tgCI and gradually decreased thereafter. On the other hand, CX3CR1 immunoreactivity was significantly increased in microglia from 5 days after tgCI. These results showed that CX3CL1 and CX3CR1 protein expression levels in pyramidal cells and microglia in the hippocampal CA1 field following tgCI were changed, indicating that tgCI-induced expression changes of CX3CL1 and CX3CR1 proteins might be closely associated with tgCI-induced delayed neuronal death and microglial activation.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyang-Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam 31116, Republic of Korea
| |
Collapse
|
28
|
Zhang M, Teng CH, Wu FF, Ge LY, Xiao J, Zhang HY, Chen DQ. Edaravone attenuates traumatic brain injury through anti-inflammatory and anti-oxidative modulation. Exp Ther Med 2019; 18:467-474. [PMID: 31281440 PMCID: PMC6580098 DOI: 10.3892/etm.2019.7632] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/11/2019] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is among the leading causes of irreversible neurological damage and death worldwide. The aim of the present study was to investigate whether edaravone (EDA) had a neuroprotective effect on TBI as well as to identify the potential mechanism. Results demonstrated that EDA suppressed inflammatory and oxidative responses in mice following TBI. This was evidenced by a reduction in glutathione peroxidase, interleukin 6, tumor necrosis factor-α and hydrogen peroxide levels, in addition to an increase in hemeoxygenase-1, quinone oxidoreductase 1 and superoxide dismutase levels, thereby mitigating neurofunctional deficits, cell apoptosis and structural damage. EDA prevented the transfer of NF-κB protein from the cytoplasm to the nucleus, whilst promoting the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) protein in mice following TBI. These results indicated that EDA exerted neuroprotective effects, including impeding neurofunctional deficits, cell apoptosis and structural damage, in mice with TBI, potentially via suppression of NF-κB-mediated inflammatory activation and promotion of the Nrf2 antioxidant pathway.
Collapse
Affiliation(s)
- Man Zhang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chen-Huai Teng
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fang-Fang Wu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Li-Yun Ge
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hong-Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Da-Qing Chen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
29
|
Zhang Y, Geng J, Hong Y, Jiao L, Li S, Sun R, Xie Y, Yan C, Aa J, Wang G. Orally Administered Crocin Protects Against Cerebral Ischemia/Reperfusion Injury Through the Metabolic Transformation of Crocetin by Gut Microbiota. Front Pharmacol 2019; 10:440. [PMID: 31114499 PMCID: PMC6502977 DOI: 10.3389/fphar.2019.00440] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 04/08/2019] [Indexed: 12/19/2022] Open
Abstract
Our pilot study suggested that orally administered crocin was hardly absorbed into circulatory system, but it was effective against cerebral ischemic/reperfusion (I/R) injury. The pharmacologically active component and targeting site of crocin remain elusive. In this study, the cerebral-protective effect of crocin was evaluated on a rat transient middle cerebral artery occlusion (MCAO) model. Our data showed that oral administration of crocin had better effectiveness in cerebral protection than an intravenous injection. Neither crocin nor its metabolite crocetin were determined in the brain of cerebral I/R rats, indicating a target site of periphery. Abundant crocetin was detected in plasma after oral administration instead of intravenous injection of crocin. Meanwhile, orally administered crocetin showed similar cerebral protection to that of crocin, but this exciting effect was not clearly observed by intravenous administration of crocetin, indicating the importance of crocetin in gut. Moreover, orally administered crocin showed less cerebral-protective effect in pseudo germ-free (pGF) MCAO rats. In vitro and in vivo experiments confirmed that crocin could be deglycosylated to crocetin in gut content of normal rats, rather than that of pGF rats, indicating that gut microbiota facilitated the transformation of crocin into crocetin, which played a key role in the activation of the pharmacological effect. Metabolomic study revealed that microbial-host co-metabolic molecules were significantly perturbed after oral administration of crocin, indicating a regulation on intestinal ecosystem. It was further suggested that gut microbiota may be the potential target of the cerebral-protective effect of crocin.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiye Aa
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | | |
Collapse
|
30
|
Fumoto T, Naraoka M, Katagai T, Li Y, Shimamura N, Ohkuma H. The Role of Oxidative Stress in Microvascular Disturbances after Experimental Subarachnoid Hemorrhage. Transl Stroke Res 2019; 10:684-694. [PMID: 30628008 DOI: 10.1007/s12975-018-0685-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/30/2018] [Accepted: 12/28/2018] [Indexed: 01/21/2023]
Abstract
Oxidative stress was shown to play a crucial role in the diverse pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Microcirculatory dysfunction is thought to be an important and fundamental pathological change in EBI. However, other than blood-brain barrier (BBB) disruption, the influence of oxidative stress on microvessels remains to be elucidated. The aim of this study was to investigate the role of oxidative stress on microcirculatory integrity in EBI. SAH was induced in male Sprague-Dawley rats using an endovascular perforation technique. A free radical scavenger, edaravone, was administered prophylactically by intraperitoneal injection. SAH grade, neurological score, brain water content, and BBB permeability were measured at 24 h after SAH induction. In addition, cortical samples taken at 24 h after SAH were analyzed to explore oxidative stress, microvascular mural cell apoptosis, microspasm, and microthrombosis. Edaravone treatment significantly ameliorated neurological deficits, brain edema, and BBB disruption. In addition, oxidative stress-induced modifications and subsequent apoptosis of microvascular endothelial cells and pericytes increased after SAH induction, while the administration of edaravone suppressed this. Consistent with apoptotic cell inhibition, microthromboses were also inhibited by edaravone administration. Oxidative stress plays a pivotal role in the induction of multiple pathological changes in microvessels in EBI. Antioxidants are potential candidates for the treatment of microvascular disturbances after SAH.
Collapse
Affiliation(s)
- Toshio Fumoto
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori, 036-8562, Japan
| | - Masato Naraoka
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori, 036-8562, Japan
| | - Takeshi Katagai
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori, 036-8562, Japan
| | - Yuchen Li
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori, 036-8562, Japan
| | - Norihito Shimamura
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori, 036-8562, Japan
| | - Hiroki Ohkuma
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori, 036-8562, Japan.
| |
Collapse
|
31
|
Li H, Tan X, Xue Q, Zhu JH, Chen G. Combined application of hypothermia and medical gases in cerebrovascular diseases. Med Gas Res 2019; 8:172-175. [PMID: 30713671 PMCID: PMC6352567 DOI: 10.4103/2045-9912.248269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/06/2018] [Indexed: 01/11/2023] Open
Abstract
Cerebrovascular diseases have a heavy burden on society and the family. At present, in the treatment of cerebrovascular diseases, the recognized effective treatment method is a thrombolytic therapy after cerebral infarction, but limited to the time window problem, many patients cannot benefit. Other treatments for cerebrovascular disease are still in the exploration stage. The study found that medical gas and hypothermia have brain protection effects. Further research found that when the two are used in combination, the therapeutic effect has a superimposed effect. This article reviews the current research progress of hypothermia therapy combined with medical gas therapy for cerebrovascular disease.
Collapse
Affiliation(s)
- Hao Li
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Tan
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qun Xue
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jue-Hua Zhu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
32
|
On-demand generation of heat and free radicals for dual cancer therapy using thermal initiator- and gold nanorod-embedded PLGA nanocomplexes. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2018.09.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
33
|
Luo P, Chu SF, Zhang Z, Xia CY, Chen NH. Fractalkine/CX3CR1 is involved in the cross-talk between neuron and glia in neurological diseases. Brain Res Bull 2018; 146:12-21. [PMID: 30496784 DOI: 10.1016/j.brainresbull.2018.11.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 11/17/2018] [Accepted: 11/23/2018] [Indexed: 01/27/2023]
Abstract
Fractalkine (CX3C chemokine ligand 1, CX3CL1) is an essential chemokine, for regulating adhesion and chemotaxis through binding to CX3CR1, which plays a critical role in the crosstalk between glial cells and neurons by direct or indirect ways in the central nervous system (CNS). Fractalkine/CX3CR1 axis regulates microglial activation and function, neuronal survival and synaptic function by controlling the release of inflammatory cytokines and synaptic plasticity in the course of the neurological disease. The multiple functions of fractalkine/CX3CR1 make it exert neuroprotective or neurotoxic effects, which determines the pathogenesis. However, the role of fractalkine/CX3CR1 in the CNS remains controversial. Whether it can be used as a therapeutic target for neurological diseases needs to be further investigated. In this review, we summarize the studies highlighting fractalkine/CX3CR1-mediated effects and discuss the potential neurotoxic and neuroprotective actions of fractalkine/CX3CR1 in brain injury for providing useful insights into the potential applications of fractalkine/CX3CR1 in neurological diseases.
Collapse
Affiliation(s)
- Piao Luo
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Cong-Yuan Xia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Nai-Hong Chen
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China.
| |
Collapse
|
34
|
Kibardina LK, Trifonov AV, Burilov AR, Pudovik MA. New Furopyridines Containing Pyridoxal and Pyrazolone Fragments. RUSS J GEN CHEM+ 2018. [DOI: 10.1134/s1070363218090098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Du S, Liu H, Lei T, Xie X, Wang H, He X, Tong R, Wang Y. Mangiferin: An effective therapeutic agent against several disorders (Review). Mol Med Rep 2018; 18:4775-4786. [PMID: 30280187 DOI: 10.3892/mmr.2018.9529] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/20/2018] [Indexed: 11/05/2022] Open
Abstract
Mangiferin (1,3,6,7‑tetrahydroxyxanthone‑C2‑β‑D‑glucoside) is a bioactive ingredient predominantly isolated from the mango tree, with potent antioxidant activity and multifactorial pharmacological effects, including antidiabetic, antitumor, lipometabolism regulating, cardioprotective, anti‑hyperuricemic, neuroprotective, antioxidant, anti‑inflammatory, antipyretic, analgesic, antibacterial, antiviral and immunomodulatory effects. Therefore, it possesses several health‑endorsing properties and is a promising candidate for further research and development. However, low solubility, mucosal permeability and bioavailability restrict the development of mangiferin as a clinical therapeutic, and chemical and physical modification is required to expand its application. This review comprehensively analyzed and collectively summarized the primary pharmacological actions of mangiferin that have been demonstrated in the literature, to support the potential future development of mangiferin as a novel therapeutic drug.
Collapse
Affiliation(s)
- Suya Du
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Huirong Liu
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Tiantian Lei
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Xiaofang Xie
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Hailian Wang
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xia He
- Personalized Drug Therapy Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Rongsheng Tong
- Personalized Drug Therapy Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yi Wang
- Personalized Drug Therapy Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
36
|
Park JH, Noh Y, Kim SS, Ahn JH, Ohk TG, Cho JH, Lee TK, Kim H, Song M, Lee JC, Won MH, Lee CH. Time-Course Changes and New Expressions of MIP-3α and Its Receptor, CCR6, in the Gerbil Hippocampal CA1 Area Following Transient Global Cerebral Ischemia. Neurochem Res 2018; 43:2102-2110. [PMID: 30203401 DOI: 10.1007/s11064-018-2632-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/13/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022]
Abstract
Macrophage inflammatory protein-3α (MIP-3α) and its sole receptor, CCR6, play pivotal roles in neuroinflammatory processes induced by brain ischemic insults. In this study, we investigated transient ischemia-induced changes in MIP-3α and CCR6 protein expressions in the hippocampal CA1 area following 5 min of transient global cerebral ischemia (tgCI) in gerbils. Both MIP-3α and CCR6 immunoreactivities were very strongly expressed in pyramidal neurons of the CA1 area from 6 h to 1 day after tgCI and were hardly shown 4 days after tgCI. In addition, strong MIP-3α immunoreactivity was newly expressed in astrocytes 6 h after tgCI. These results indicate that tgCI causes apparent changes in MIP-3α and CCR6 expressions in pyramidal neurons and astrocytes in the hippocampal CA1 area and suggest that tgCI-induced changes in MIP-3α and CCR6 expressions might be closely associated with neuroinflammatory processes in brain ischemic regions.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Republic of Korea
| | - YooHun Noh
- Famenity Company, Gwacheon, Geyonggi, 13837, Republic of Korea
| | - Sung-Su Kim
- Famenity Company, Gwacheon, Geyonggi, 13837, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea.
| |
Collapse
|
37
|
Landreneau MJ, Mullen MT, Messé SR, Cucchiara B, Sheth KN, McCullough LD, Kasner SE, Sansing LH. CCL2 and CXCL10 are associated with poor outcome after intracerebral hemorrhage. Ann Clin Transl Neurol 2018; 5:962-970. [PMID: 30128320 PMCID: PMC6093844 DOI: 10.1002/acn3.595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/02/2018] [Accepted: 05/22/2018] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Intracerebral hemorrhage carries a high mortality and survivors are frequently left with significant disability. Immunological mechanisms may play an important role in hemorrhage-induced brain injury, however, research linking these mechanisms with clinical outcome remains limited. We aim to identify serum inflammatory mediators that are associated with outcome after intracerebral hemorrhage in order to translate data from experimental models to a patient cohort and identify potential targets worthy of reverse translation. METHODS A prospective cohort study at two comprehensive stroke centers enrolled patients with spontaneous intracerebral hemorrhage. Peripheral blood was collected at 6, 24, and 72 h from onset. Functional outcome was assessed at 90 days using the modified Rankin Scale (mRS). Serum inflammatory mediators were measured using multiplex ELISA. Multivariable modeling identified serum biomarkers independently associated with functional outcome at 90 days. RESULTS 115 patients completed the study. At 6 h after onset, patients with elevated CCL2 had worse mRS score at day 90 (OR 4.07, 95% CI 1.27-13.10, P = 0.02) after adjusting for age, gender, ICH volume, IVH, infratentorial location and NIHSS score. At 24 and 72 h after onset, elevation in CXCL10 was independently associated with worse 90 days mRS score (24 h: OR 8.08, 95% CI 2.69-24.30, P < 0.001; 72 h: OR 3.89, 95% CI 1.12-13.49, P = 0.03). INTERPRETATION Acute and subacute elevations in specific immune factors are associated with poor outcome, highlighting potential pathways that may contribute to ongoing brain injury in patients with intracerebral hemorrhage.
Collapse
Affiliation(s)
| | - Michael T. Mullen
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Steven R. Messé
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Brett Cucchiara
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Kevin N. Sheth
- Department of NeurologyYale University School of MedicineNew HavenConnecticut
- Center for Neuroepidemiology and Clinical Neurological ResearchYale School of MedicineNew HavenConnecticut
| | - Louise D. McCullough
- Department of NeurologyUniversity of Texas Health Sciences Center at HoustonHoustonTexas
| | - Scott E. Kasner
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Lauren H. Sansing
- Department of NeurologyYale University School of MedicineNew HavenConnecticut
- Center for Neuroepidemiology and Clinical Neurological ResearchYale School of MedicineNew HavenConnecticut
| | | |
Collapse
|
38
|
Takase H, Liang AC, Miyamoto N, Hamanaka G, Ohtomo R, Maki T, Pham LDD, Lok J, Lo EH, Arai K. Protective effects of a radical scavenger edaravone on oligodendrocyte precursor cells against oxidative stress. Neurosci Lett 2018; 668:120-125. [PMID: 29337010 PMCID: PMC5829007 DOI: 10.1016/j.neulet.2018.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/26/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) play critical roles in maintaining the number of oligodendrocytes in white matter. Previously, we have shown that oxidative stress dampens oligodendrocyte regeneration after white matter damage, while a clinically proven radical scavenger, edaravone, supports oligodendrocyte repopulation. However, it is not known how edaravone exerts this beneficial effect against oxidative stress. Using in vivo and in vitro experiments, we have examined whether edaravone exhibits direct OPC-protective effects. For in vivo experiments, prolonged cerebral hypoperfusion was induced by bilateral common carotid artery stenosis in mice. OPC damage was observed on day 14 after the onset of cerebral hypoperfusion, and edaravone was demonstrated to decrease OPC death in cerebral white matter. In vitro experiments also confirmed that edaravone reduced oxidative-stress-induced OPC death. Because white matter damage is a major hallmark of many neurological diseases, and OPCs are instrumental in white matter repair after injury, our current study supports the idea that radical scavengers may provide a potential therapeutic approach for white matter related diseases.
Collapse
Affiliation(s)
- Hajime Takase
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA; Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Japan
| | - Anna C Liang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Nobukazu Miyamoto
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Ryo Ohtomo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Loc-Duyen D Pham
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA; Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA.
| |
Collapse
|
39
|
Mizuma A, Yenari MA. Anti-Inflammatory Targets for the Treatment of Reperfusion Injury in Stroke. Front Neurol 2017; 8:467. [PMID: 28936196 PMCID: PMC5594066 DOI: 10.3389/fneur.2017.00467] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
While the mainstay of acute stroke treatment includes revascularization via recombinant tissue plasminogen activator or mechanical thrombectomy, only a minority of stroke patients are eligible for treatment, as delayed treatment can lead to worsened outcome. This worsened outcome at the experimental level has been attributed to an entity known as reperfusion injury (R/I). R/I is occurred when revascularization is delayed after critical brain and vascular injury has occurred, so that when oxygenated blood is restored, ischemic damage is increased, rather than decreased. R/I can increase lesion size and also worsen blood barrier breakdown and lead to brain edema and hemorrhage. A major mechanism underlying R/I is that of poststroke inflammation. The poststroke immune response consists of the aberrant activation of glial cell, infiltration of peripheral leukocytes, and the release of damage-associated molecular pattern (DAMP) molecules elaborated by ischemic cells of the brain. Inflammatory mediators involved in this response include cytokines, chemokines, adhesion molecules, and several immune molecule effectors such as matrix metalloproteinases-9, inducible nitric oxide synthase, nitric oxide, and reactive oxygen species. Several experimental studies over the years have characterized these molecules and have shown that their inhibition improves neurological outcome. Yet, numerous clinical studies failed to demonstrate any positive outcomes in stroke patients. However, many of these clinical trials were carried out before the routine use of revascularization therapies. In this review, we cover mechanisms of inflammation involved in R/I, therapeutic targets, and relevant experimental and clinical studies, which might stimulate renewed interest in designing clinical trials to specifically target R/I. We propose that by targeting anti-inflammatory targets in R/I as a combined therapy, it may be possible to further improve outcomes from pharmacological thrombolysis or mechanical thrombectomy.
Collapse
Affiliation(s)
- Atsushi Mizuma
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Midori A Yenari
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA, United States
| |
Collapse
|
40
|
Zhang J, Liu R, Kuang HY, Gao XY, Liu HL. Protective treatments and their target retinal ganglion cells in diabetic retinopathy. Brain Res Bull 2017; 132:53-60. [DOI: 10.1016/j.brainresbull.2017.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 05/10/2017] [Indexed: 12/19/2022]
|
41
|
Zhang CM, Qin SY, Cheng YJ, Zhang AQ. Construction of poly(dopamine) doped oligopeptide hydrogel. RSC Adv 2017. [DOI: 10.1039/c7ra10363h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Incorporation of poly(dopamine) (PDA) into the native oligopeptide hydrogel not only improve the rigidity but also endow the hydrogel with efficient free radical scavenging ability.
Collapse
Affiliation(s)
- Chen-Ming Zhang
- College of Chemistry and Materials Science
- South-Central University for Nationalities
- Wuhan 430074
- P. R. China
| | - Si-Yong Qin
- College of Chemistry and Materials Science
- South-Central University for Nationalities
- Wuhan 430074
- P. R. China
| | - Yin-Jia Cheng
- College of Chemistry and Materials Science
- South-Central University for Nationalities
- Wuhan 430074
- P. R. China
| | - Ai-Qing Zhang
- College of Chemistry and Materials Science
- South-Central University for Nationalities
- Wuhan 430074
- P. R. China
| |
Collapse
|
42
|
Bonaventura A, Liberale L, Vecchié A, Casula M, Carbone F, Dallegri F, Montecucco F. Update on Inflammatory Biomarkers and Treatments in Ischemic Stroke. Int J Mol Sci 2016; 17:1967. [PMID: 27898011 PMCID: PMC5187767 DOI: 10.3390/ijms17121967] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/08/2016] [Accepted: 11/17/2016] [Indexed: 12/26/2022] Open
Abstract
After an acute ischemic stroke (AIS), inflammatory processes are able to concomitantly induce both beneficial and detrimental effects. In this narrative review, we updated evidence on the inflammatory pathways and mediators that are investigated as promising therapeutic targets. We searched for papers on PubMed and MEDLINE up to August 2016. The terms searched alone or in combination were: ischemic stroke, inflammation, oxidative stress, ischemia reperfusion, innate immunity, adaptive immunity, autoimmunity. Inflammation in AIS is characterized by a storm of cytokines, chemokines, and Damage-Associated Molecular Patterns (DAMPs) released by several cells contributing to exacerbate the tissue injury both in the acute and reparative phases. Interestingly, many biomarkers have been studied, but none of these reflected the complexity of systemic immune response. Reperfusion therapies showed a good efficacy in the recovery after an AIS. New therapies appear promising both in pre-clinical and clinical studies, but still need more detailed studies to be translated in the ordinary clinical practice. In spite of clinical progresses, no beneficial long-term interventions targeting inflammation are currently available. Our knowledge about cells, biomarkers, and inflammatory markers is growing and is hoped to better evaluate the impact of new treatments, such as monoclonal antibodies and cell-based therapies.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Alessandra Vecchié
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Matteo Casula
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genova, 10 Largo Benzi, 16132 Genoa, Italy.
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genova, 10 Largo Benzi, 16132 Genoa, Italy.
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|