1
|
Ma Y, Hu X, Shen S, Pan D. Geniposide ameliorates brain injury in mice with intracerebral hemorrhage by inhibiting NF-κB signaling. Neurol Res 2024; 46:346-355. [PMID: 38402902 DOI: 10.1080/01616412.2024.2321014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Neuroinflammation and oxidative stress are critical players in intracerebral hemorrhage (ICH). Geniposide is an active component of Gardenia that has anti-inflammatory effects. This study focused on the roles and mechanisms of geniposide in ICH. METHODS ICH was established by injecting collagenase IV into C57BL/6 mice. To determine the functions of geniposide and NF-κB inhibition in ICH model mice, geniposide (1, 25, or 50 mg/kg) or PDTC (a NF-κB inhibitor) was administered. Neurological functions were assessed with the modified neurological severity score (mNSS) test. Hematoxylin and eosin staining were performed to identify pathological changes. IL-1β and TNF-α levels were estimated with ELISA kits. NF-κB p65 localization was determined by immunofluorescence staining. Oxidative stress was analyzed by measuring ROS levels. RESULTS Geniposide alleviated cerebral edema and neurological deficits. Geniposide inhibited neuroinflammation and oxidative stress after ICH, and the inhibitory effects were enhanced by NF-κB inhibition. Additionally, geniposide inhibited NF-κB signaling. CONCLUSION Geniposide alleviates brain injury by suppressing inflammation and oxidative stress damage in experimental ICH models by inhibiting NF-κB signaling.
Collapse
Affiliation(s)
- Yinghui Ma
- Department of Neurosurgery, Huang Shi Central Hospital, HuangShi, China
| | - Xiao Hu
- Department of Neurosurgery, Huang Shi Central Hospital, HuangShi, China
| | - Songbo Shen
- Department of Neurosurgery, Huang Shi Central Hospital, HuangShi, China
| | - Dongmei Pan
- Department of Geriatrics, Huang Shi Central Hospital, HuangShi, China
| |
Collapse
|
2
|
Yin H, Ran Z, Luo T, Jin Z, Ma J. BCL-3 Promotes Intracerebral Hemorrhage Progression by Increasing Blood-Brain Barrier Permeability, Inflammation, and Cell Apoptosis via Endoplasmic Reticulum Stress. Mediators Inflamm 2023; 2023:1420367. [PMID: 37736616 PMCID: PMC10511295 DOI: 10.1155/2023/1420367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 09/23/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH) is among the common types of stroke with high mortality and morbidity. Molecular biomarker selection is crucial for ICH diagnosis and treatment. However, the identification of ICH-related biomarkers remains inadequate. Materials and Methods In vivo and in vitro ICH models were generated and transfected with silenced B-cell lymphoma-3 (BCL-3 and siRNA BCL-3), overexpressed BCL-3, and endoplasmic reticulum stress (ERS) agonist (2-CLHA). Hematoxylin-eosin staining and transmission electron microscopy were used to observe the transfected cells. RNA sequencing was performed in vivo on the sham and ICH groups. The blood-brain barrier (BBB) permeability was evaluated by determining Evans blue dye extravasation, transendothelial electrical resistance, and paracellular permeability. Moreover, tight junction-, cell apoptosis-, and endoplasmic reticulum stress- (ERS-) related proteins were evaluated through real-time quantitative PCR, western blotting, immunohistochemistry, and TUNEL staining. The levels of inflammatory cytokines were measured through the enzyme-linked immunosorbent assay. Results RNA-seq revealed that BCL-3 acts as a key player. BCL-3 promotes ICH progression by increasing BBB permeability, ERS, inflammation, and cell apoptosis. Silencing of BCL-3 slows ICH progression by reducing BBB permeability and inflammation and terminating cell apoptosis and ERS in vitro and in vivo. Conclusion Our study identified ICH biomarkers and elucidated the role of BCL-3 in ICH for the first time.
Collapse
Affiliation(s)
- Hao Yin
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Zhongying Ran
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Tao Luo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Zexin Jin
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Jun Ma
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| |
Collapse
|
3
|
Nakanishi S, Kinoshita K, Kurauchi Y, Seki T, Kimura Y, Suzuki M, Suzuki K, Koyama H, Kagechika H, Katsuki H. Acyclic retinoid peretinoin reduces hemorrhage-associated brain injury in vitro and in vivo. Eur J Pharmacol 2023; 954:175899. [PMID: 37392831 DOI: 10.1016/j.ejphar.2023.175899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/03/2023]
Abstract
Peretinoin is an acyclic retinoid that stimulates retinoic acid receptors (NR1Bs) and produces therapeutic effects on hepatocellular cancer. We have previously shown that NR1B agonists such as Am80 and all trans-retinoic acid suppress pathogenic events in intracerebral hemorrhage. The present study addressed the actions of peretinoin and Am80 against cytotoxicity of a blood protease thrombin on cortico-striatal slice cultures obtained from neonatal rat brains. Application of 100 U/ml thrombin to the slice cultures for 72 h caused cell death in the cortical region and tissue shrinkage in the striatal region. Peretinoin (50 μM) and Am80 (1 μM) counteracted these cytotoxic effects of thrombin, and the effect of peretinoin and Am80 was blocked by LE540, an NR1B antagonist. A broad-spectrum kinase inhibitor K252a (3 μM) attenuated the cytoprotective effect of peretinoin in the cortical region, whereas a specific protein kinase A inhibitor KT5720 (1 μM) attenuated the protective effect of peretinoin in the cortical and the striatal regions. On the other hand, nuclear factor-κB (NF-κB) inhibitors such as pyrrolidine dithiocarbamate (50 μM) and Bay11-7082 (10 μM) prevented thrombin-induced shrinkage of the striatal region. Peretinoin and Am80 as well as Bay11-7082 blocked thrombin-induced nuclear translocation of NF-κB in striatal microglia and loss of striatal neurons. We also found that daily administration of peretinoin reduced histopathological injury and alleviated motor deficits in a mouse model of intracerebral hemorrhage. These results indicate that NR1B agonists including peretinoin may serve as a therapeutic option for hemorrhagic brain injury.
Collapse
Affiliation(s)
- Sakino Nakanishi
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keita Kinoshita
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahiro Seki
- Department of Pharmacology, School of Pharmacy, Himeji-Dokkyo University, Hyogo, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Masaaki Suzuki
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Keiichi Suzuki
- Field of Biological Molecular Sciences, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Hiroko Koyama
- Field of Biological Molecular Sciences, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan; Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
4
|
Xu Y, Li Y, Wang C, Han T, Liu H, Sun L, Hong J, Hashimoto M, Wei J. The reciprocal interactions between microglia and T cells in Parkinson's disease: a double-edged sword. J Neuroinflammation 2023; 20:33. [PMID: 36774485 PMCID: PMC9922470 DOI: 10.1186/s12974-023-02723-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/08/2023] [Indexed: 02/13/2023] Open
Abstract
In Parkinson's disease (PD), neurotoxic microglia, Th1 cells, and Th17 cells are overactivated. Overactivation of these immune cells exacerbates the disease process and leads to the pathological development of pro-inflammatory cytokines, chemokines, and contact-killing compounds, causing the loss of dopaminergic neurons. So far, we have mainly focused on the role of the specific class of immune cells in PD while neglecting the impact of interactions among immune cells on the disease. Therefore, this review demonstrates the reciprocal interplays between microglia and T cells and the associated subpopulations through cytokine and chemokine production that impair and/or protect the pathological process of PD. Furthermore, potential targets and models of PD neuroinflammation are highlighted to provide the new ideas/directions for future research.
Collapse
Affiliation(s)
- Yuxiang Xu
- grid.256922.80000 0000 9139 560XInstitute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004 China ,grid.256922.80000 0000 9139 560XHenan International Joint Laboratory for Nuclear Protein Regulation, Henan Medical School, Henan University, Kaifeng, 475004 China
| | - Yongjie Li
- grid.414360.40000 0004 0605 7104Department of Rehabilitation Medicine, Beijing Jishuitan Hospital Guizhou Hospital, Guizhou Provincial Orthopedics Hospital, Guiyang, China
| | - Changqing Wang
- grid.256922.80000 0000 9139 560XInstitute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004 China
| | - Tingting Han
- grid.256922.80000 0000 9139 560XInstitute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004 China
| | - Haixuan Liu
- grid.256922.80000 0000 9139 560XInstitute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004 China
| | - Lin Sun
- grid.256922.80000 0000 9139 560XHenan Key Laboratory of Polyoxometalate Chemistry, College of Chemistry and Chemical Engineering, Henan University, Kaifeng, 475004 Henan China
| | - Jun Hong
- grid.256922.80000 0000 9139 560XInstitute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004 China
| | - Makoto Hashimoto
- grid.272456.00000 0000 9343 3630Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506 Japan
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China. .,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan Medical School, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
5
|
Yu M, Tian T, Zhang J, Hu T. miR-141-3p protects against blood-brain barrier disruption and brain injury after intracerebral hemorrhage by targeting ZEB2. J Clin Neurosci 2022; 99:253-260. [PMID: 35306455 DOI: 10.1016/j.jocn.2022.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/28/2022] [Accepted: 03/05/2022] [Indexed: 12/31/2022]
Abstract
MicroRNAs (miRNAs) participate in the diagnosis and treatment of intracerebral hemorrhage (ICH). miR-141-3p has been widely reported to regulate neurological disorders and cerebropathy. However, the specific role of miR-141-3p in ICH has not yet been revealed. The aim of this study was exploration of the biological functions and mechanism of miR-141-3p in ICH by establishing a collagenase-induced ICH mouse model. After ICH induction, miR-141-3p mimics or miR-NC were administered into the right striatum of the model mice followed by the performance of neurological tests. After euthanasia of the mice, the injury volume, brain water content, and injury to the blood-brain barrier (BBB) were evaluated. Evans blue (EB) was used to stain the brain slices, and EB extravasation was detected to evaluate the injury to BBB. miR-141-3p expression in perihematomal edema and hematoma areas after ICH was assessed by RT-qPCR. The levels of tight junction proteins in brain tissues and human brain microvascular endothelial cells (BMECs) were evaluated by western blotting. The FITC-dextran 20 method was used to assess BMEC permeability. The binding between miR-141-3p and zinc finger E-box-binding homeobox 2 (ZEB2) was verified with a luciferase reporter assay. In this study, miR-141-3p overexpression alleviated ICH-induced brain injury and protected BBB integrity in vivo. ZEB2 was a target gene of miR-141-3p. ZEB2 overexpression promoted BBB disruption, and miR-141-3p overexpression attenuated the promoting effect exerted by ZEB2. Overall, miR-141-3p protects against BBB disruption and attenuates brain injuries induced by ICH by targeting ZEB2.
Collapse
Affiliation(s)
- Miao Yu
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Tian Tian
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China.
| | - Jiwei Zhang
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Tiemin Hu
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| |
Collapse
|
6
|
WANG FD, LI J, ZHAI X, CHEN R, WANG F. Methane-rich saline restores brain SOD activity and alleviates cognitive impairment in rats with traumatic brain injury. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.54921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Jie LI
- Xi'an Jiaotong University, China
| | - Xu ZHAI
- Xi'an Jiaotong University, China
| | - Rui CHEN
- Xi'an Jiaotong University, China
| | | |
Collapse
|
7
|
Anti-CD47 antibody administration via cisterna magna in proper dosage can reduce perihematomal cell death following intracerebral hemorrhage in rats. Brain Res Bull 2021; 174:359-365. [PMID: 34252444 DOI: 10.1016/j.brainresbull.2021.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The secondary injury caused by RBC autolysis after intracerebral hemorrhage (ICH) can be reduced by increasing the efficiency of microglia (MG)/macrophages (Mø) phagocytizing red blood cells (RBCs). CD47 is an important regulator of MG/Mø phagocytosis. This study aims to clarify whether anti-CD47 antibody administrated into the cisterna magna after ICH can transfer to the hematoma site, promote MG/Mø gathering to phagocytize RBCs and ultimately reduce cell death. METHODS Forty male Wistar rats were divided into sham, ICH, low-dosage (group A, 0.3 μg), medium-dosage (group B, 0.9 μg) and high-dosage (group C, 1.8 μg) anti-CD47 antibody groups. For the rats in group A, B and C, anti-CD47 antibody solution was administrated into the cisterna magna at 10 min after ICH. Brain tissue was harvested 3 days after the operation. Western blotting was performed to detect the expression of Caspase-3 and Bcl-2. Immunofluorescence was performed to detect the CD68 expression. TUNEL was performed to detect the cell death. RESULTS The hematoma of the ICH rats was located in the basal ganglia, with a good homogeneity of hematoma volume. Low-dosage anti-CD47 antibody in group A had no effects on the perihematomal CD68 (P = 0.338), Caspase-3 (P = 0.769), Bcl-2 (P = 0.176) expression and cell death (P = 0.698), compared with the ICH group. CD68 and Bcl-2 expression increased and Caspase-3 expression decreased significantly in group B (P < 0.001 for all) and group C (P < 0.001 for all). The increase of CD68 expression in group C was greater than that in group B (P < 0.01) by a large margin, while there was no difference for Bcl-2 (P = 0.908) and Caspase-3 (P = 0.913) expression between the 2 groups. Compared with the ICH group, medium-dosage of anti-CD47 antibody in group B significantly reduced the number of TUNEL-positive cells (P < 0.005), but not for group C (P = 0.311). CONCLUSION The results suggested that anti-CD47 antibody administration into the cisterna magna in proper dosage (0.9 μg) can effectively reach the hematoma, induce more MG/Møs to gather around the hematoma, and reduce cell death in perihematomal brain tissue. The results of this study has provided a basic theory for improving the efficiency of MG/Mø phagocytizing RBCs and hematoma clearance after ICH by administrating anti-CD47 antibody via the cisterna magna.
Collapse
|
8
|
Deng S, Jin P, Sherchan P, Liu S, Cui Y, Huang L, Zhang JH, Gong Y, Tang J. Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice. J Neuroinflammation 2021; 18:62. [PMID: 33648537 PMCID: PMC7923481 DOI: 10.1186/s12974-021-02112-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance after ICH. However, it is unclear whether CCR4 activation can ameliorate neuroinflammation and apoptosis of neurons following ICH. The aim of the present study was to examine the effects of recombinant CCL17 (rCCL17)-dependent CCR4 activation on neuroinflammation and neuronal apoptosis in an intrastriatal autologous blood injection ICH model, and to determine whether the PI3K/AKT/Foxo1 signaling pathway was involved. Methods Two hundred twenty-six adult (8-week-old) male CD1 mice were randomly assigned to sham and ICH surgery groups. An intrastriatal autologous blood injection ICH model was used. rCCL17, a CCR4 ligand, was delivered by intranasal administration at 1 h, 3 h, and 6 h post-ICH. CCL17 antibody was administrated by intraventricular injection at 1 h post-ICH. C021, a specific inhibitor of CCR4 and GDC0068, an AKT inhibitor were delivered intraperitoneally 1 h prior to ICH induction. Brain edema, neurobehavioral assessments, western blotting, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunofluorescence staining were conducted. Results Endogenous expression of CCL17 and CCR4 were increased following ICH, peaking at 5 days post-induction. CCR4 was found to co-localize with microglia, neurons, and astrocytes. rCCL17 treatment decreased brain water content, attenuated short- and long-term neurological deficits, deceased activation of microglia/macrophages and infiltration of neutrophils, and inhibited neuronal apoptosis in the perihematomal region post-ICH. Moreover, rCCL17 treatment post-ICH significantly increased the expression of CCR4, PI3K, phosphorylated AKT, and Bcl-2, while Foxo1, IL-1β, TNF-α, and Bax expression were decreased. The neuroprotective effects of rCCL17 were reversed with the administration of C021 or GDC0068. Conclusions rCCL17-dependent CCR4 activation ameliorated neurological deficits, reduced brain edema, and ameliorated neuroinflammation and neuronal apoptosis, at least in part, through the PI3K/AKT/Foxo1 signaling pathway after ICH. Thus, activation of CCR4 may provide a promising therapeutic approach for the early management of ICH. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02112-3.
Collapse
Affiliation(s)
- Shuixiang Deng
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, 12 middle WuLuMuQi, Shanghai, 200040, China.,Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Peng Jin
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, 12 middle WuLuMuQi, Shanghai, 200040, China.,Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Shengpeng Liu
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, China
| | - Yuhui Cui
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Ye Gong
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, 12 middle WuLuMuQi, Shanghai, 200040, China. .,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.
| |
Collapse
|
9
|
Yang M, Feng Y, Yan S, Wu Z, Xiao X, Sang J, Ye S, Liu F, Cui W. Evans Blue Might Produce Pathologically Activated Neuroprotective Effects via the Inhibition of the P2X4R/p38 Signaling Pathway. Cell Mol Neurobiol 2021; 41:293-307. [PMID: 32382851 DOI: 10.1007/s10571-020-00852-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
The main pathological features of ischemic stroke include neuronal damage and blood-brain barrier (BBB) dysfunction. Previous studies have shown that Evans Blue, a dye used to probe BBB integrity, could enter the brain only during the pathological status of ischemic stroke, indicating the potential pathologically activated therapeutic use of this chemical to treat ischemic stroke. In this study, we have reported that Evans Blue could produce in vitro neuroprotective effects against iodoacetic acid (IAA)-induced hypoxia neuronal death in HT22 cells. We further found that P2X purinoreceptor 4 (P2X4R), a subtype of ATP-gated cation channel, was expressed in HT22 cells. Evans Blue could prevent IAA-induced increase of P2X4R mRNA and protein expression. Interestingly, shRNA of P2X4R could protect against IAA-induced activation of p38, and SB203580, a specific inhibitor of p38, could reverse IAA-induced neurotoxicity, indicating that p38 is a downstream signaling molecule of P2X4R. Molecular docking analysis further demonstrated the possible interaction between Evans Blue and the ATP binding site of P2X4R. Most importantly, pre-treatment of Evans Blue could largely reduce neurological and behavioral abnormity, and decrease brain infarct volume in middle cerebral artery occlusion/reperfusion (MCAO) rats. All these results strongly suggested that Evans Blue could exert neuroprotective effects via inhibiting the P2X4R/p38 pathway, possibly by acting on the ATP binding site of P2X4R, indicating that Evans Blue might be further developed as a pathologically activated therapeutic drug against ischemic stroke.
Collapse
Affiliation(s)
- Mengxiang Yang
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Yi Feng
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Sicheng Yan
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Zhuoying Wu
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Xiao Xiao
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Jingcheng Sang
- Key Laboratory of Industrial Fermentation Microbiology of Education, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shazhou Ye
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology of Education, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China.
- Department of Physiology, Medical School, Ningbo University, Zhejiang, China.
| |
Collapse
|
10
|
Zhou W, Huang G, Ye J, Jiang J, Xu Q. Protective Effect of miR-340-5p against Brain Injury after Intracerebral Hemorrhage by Targeting PDCD4. Cerebrovasc Dis 2020; 49:593-600. [PMID: 33176298 DOI: 10.1159/000508210] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/25/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Intracerebral hemorrhage (ICH) is a common cerebrovascular disease. Increasing evidence has documented the crucial role of microRNAs in ICH. The present study aimed to investigate the role and underlying mechanism of miR-340-5p in ICH. METHODS The collagenase-induced ICH rat model was established. The neurological function of rats and the cerebral water content of rat brain tissue were measured to assess the brain injury. BV-2 cells were recruited and treated by LPS to mimic ICH-induced inflammatory response. qRT-PCR was used for the measurement of miR-340-5p. The protein levels of TNF-α, IL-6, and IL-1β were detected using ELISA. Luciferase reporter gene assay was performed to confirm the target gene. RESULTS Downregulation of miR-340-5p was detected in the serum of ICH patients and the brain tissues of ICH rats. Overexpression of miR-340-5p reversed the influence of ICH on the neurological function score and cerebral water content and inhibited the production of proinflammatory cytokines (TNF-α, IL-6, and IL-1β), which were induced by ICH in vivo. In in vitro study, levels of TNF-α, IL-6, and IL-1β were significantly enhanced in cells after LPS treatment, but these increases were eliminated by overexpression of miR-340-5p. PDCD4 was a direct target gene of miR-340-5p. CONCLUSION miR-340-5p protects against brain injury after ICH. miR-340-5p might exert an anti-inflammatory effect during the occurrence of ICH via targeting PDCD4.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, Shanghai, China
| | - Guandong Huang
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, Shanghai, China
| | - Jueming Ye
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, Shanghai, China
| | - Jiamei Jiang
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, Shanghai, China,
| | - Qing Xu
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, Shanghai, China
| |
Collapse
|