101
|
Amino Acid Biosignature in Plasma among Ischemic Stroke Subtypes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8480468. [PMID: 30800679 PMCID: PMC6360633 DOI: 10.1155/2019/8480468] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/29/2018] [Accepted: 12/10/2018] [Indexed: 12/28/2022]
Abstract
Ischemic stroke is a neurovascular disorder caused by reduced or blockage of blood flow to the brain, which may permanently affect motor and cognitive abilities. The diagnostic of stroke is performed using imaging technologies, clinical evaluation, and neuropsychological protocols, but no blood test is available yet. In this work, we analyzed amino acid concentrations in blood plasma from poststroke patients in order to identify differences that could characterize the stroke etiology. Plasma concentrations of sixteen amino acids from patients with chronic ischemic stroke (n = 73) and the control group (n = 16) were determined using gas chromatography coupled to mass spectrometry (GC-MS). The concentration data was processed by Partial Least Squares-Discriminant Analysis (PLS-DA) to classify patients with stroke and control. The amino acid analysis generated a first model able to discriminate ischemic stroke patients from control group. Proline was the most important amino acid for classification of the stroke samples in PLS-DA, followed by lysine, phenylalanine, leucine, and glycine, and while higher levels of methionine and alanine were mostly related to the control samples. The second model was able to discriminate the stroke subtypes like atherothrombotic etiology from cardioembolic and lacunar etiologies, with lysine, leucine, and cysteine plasmatic concentrations being the most important metabolites. Our results suggest an amino acid biosignature for patients with chronic stroke in plasma samples, which can be helpful in diagnosis, prognosis, and therapeutics of these patients.
Collapse
|
102
|
Viswanatha GL, Venkataranganna MV, Prasad NBL, Shylaja H. Achyranthes aspera Linn. alleviates cerebral ischemia-reperfusion-induced neurocognitive, biochemical, morphological and histological alterations in Wistar rats. JOURNAL OF ETHNOPHARMACOLOGY 2019; 228:58-69. [PMID: 30223049 DOI: 10.1016/j.jep.2018.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/04/2018] [Accepted: 09/11/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the traditional system of Indian medicine, the whole plant and roots of Achyranthes aspera L have been extensively used to treat neurological conditions such as epilepsy and stroke by the various ethnic communities of India. AIM OF THE STUDY The present study was aimed to evaluate the cerebroprotective potential of methanol extract of A. aspera aerial parts (MeAA). MATERIALS AND METHODS Initially the MeAA was evaluated for total phenolic content and subjected to detailed liquid chromatography-mass spectrometry analysis. Additionally, it was evaluated for in vitro antioxidant activity in ferric reducing antioxidant power, 2, 2-diphenyl-1-picrylhydrazyl and oxygen radical absorbance capacity assays. Furthermore, in RAW 264.7 cell lines the effect of MeAA was evaluated on lipopolysaccharide-induced generation of reactive oxygen species, nitrite and tumor necrosis factor-α. Finally, the MeAA (400 and 800 mg/kg) was evaluated against ischemia-reperfusion (I/R)-induced brain injury in rats. In brief, male Wistar rats were allocated in to five groups (G-I to G-V, n = 10). G-I and G-II assigned as sham control and I/R control, and received only vehicle (carboxy methyl cellulose 0.5% w/v, 10 ml/kg, p.o.). G-III received quercetin (20 mg/kg, p.o.) and assigned as reference standard. G-IV and G-V group animals received 400 and 800 mg/kg oral doses of MeAA, respectively. All the treatments were given orally for a period of seven days and the parameters such as functional (neurological, cognitive and motor), morphological (edema and infarct area), biochemical (superoxide dismutase, catalase, reduced glutathione, lipid peroxidation, cytokines), and histopathological evaluations of the brain tissue was performed. RESULTS The MeAA exhibited 72.48 mg gallic acid equivalents/g of total phenolic content and the LC-MS/MS analysis showed acteoside, apigenin, and pentagalloyl glucose as major ingredients in the MeAA. In in vitro antioxidant assays, the MeAA showed good antioxidant activity with IC50 of 126.50 μg/ml in DPPH assay; FRAP and ORAC values of 759.65 and 979.4 in FRAP and ORAC assays, respectively. Further, the MeAA significantly suppressed the generation of ROS, nitrite and TNF-α in LPS activated RAW 264.7 cell lines. Besides, sixty mins of global cerebral ischemia followed by 24 h of reperfusion produced considerable alterations in neurobehavioral functions in the I/R control group compared to sham control, with a significant reduction in catalase and superoxide dismutase enzyme activities. Moreover, there was a significant reduction in reduced glutathione levels with increased lipid peroxidation. Furthermore, the levels of pro-inflammatory cytokines (TNF-α, IL-6, and ICAM-I) increased significantly and those of anti-inflammatory (IL-10) decreased. I/R insult increased the brain volume and aggravated cerebral infarct formation. Histopathological examination of the brain tissue revealed vascular congestion, cerebral edema, leukocyte infiltration, and brain tissue necrosis. Interestingly, seven days pretreatment with MeAA (800 mg/kg, p.o.) has offered significant protection against I/R-induced functional, morphological, biochemical and histopathological alterations in Wistar rats. CONCLUSIONS These findings suggest that the MeAA possesses potent cerebroprotective action through its antioxidant and anti-inflammatory mechanisms.
Collapse
|
103
|
Wang CW, Ma PJ, Wang YY, Yang M, Su LL, Wang S, Liu YX, Yuan B, Zhao JH. Serum level of macrophage migration inhibitory factor predicts severity and prognosis in patients with ischemic stroke. Cytokine 2019; 115:8-12. [PMID: 30616035 DOI: 10.1016/j.cyto.2018.11.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/28/2018] [Accepted: 11/25/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To evaluate whether the macrophage migration inhibitory factor (MIF) level in serum of ischemic stroke patients was associated with their clinical severity and early outcome. METHODS During February 2017-March 2018, consecutive patients admitted to our hospital because of first-ever ischemic stroke were identified. The prognostic value of MIF was set for predicting the outcome of these patients at discharge. The results were compared with existing methods, including National Institutes of Health Stroke Scale (NIHSS) score and validated indicators. RESULTS 289 patients were enrolled. The serum level of all patients was determined (median: 20.6 ng/ml). At admission, 131 patients (45.3%) were evaluated as minor stroke (NIHSS < 5). When serum level of MIF was increased by each 1 ng/ml, the unadjusted and adjusted risk of moderate-to-high clinical severity was elevated by 5% (OR = 1.05 [95% CI: 1.01-1.09], P = 0.006) and 3% (1.03 [1.00-1.08], P = 0.02), respectively. At discharge, 82 patients (28.4%) had poor functional outcomes. The median serum level of MIF was lower in group with good outcomes than that observed in poor outcomes (19.4[15.8-24.2] vs. 24.0[19.9-29.4] ng/ml; P < 0.001). When serum level of MIF was increased by each 1 ng/ml, the unadjusted and adjusted risk of poor outcomes was elevated by 9% (1.09 [1.05-1.13], P < 0.001) and 6% (1.06 [1.02-1.10], P < 0.01), respectively. CONCLUSIONS High MIF levels are independently related to the moderate to high clinical severity in ischemic stroke patients, as well as the poor outcome at discharge.
Collapse
Affiliation(s)
- Chao-Wei Wang
- Department of Neurology II, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Henan Key Laboratory of Neural Regeneration, Xinxiang, China
| | - Peng-Ju Ma
- Henan Key Laboratory of Neural Regeneration, Xinxiang, China; Department of Neurosurgery I, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yang-Yang Wang
- Henan Key Laboratory of Neural Regeneration, Xinxiang, China; Department of Neurosurgery I, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ming Yang
- Henan Key Laboratory of Neural Regeneration, Xinxiang, China; Department of Neurosurgery I, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Lin-Lin Su
- Department of Neurology II, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Henan Key Laboratory of Neural Regeneration, Xinxiang, China
| | - Shuo Wang
- Department of Neurology II, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Henan Key Laboratory of Neural Regeneration, Xinxiang, China
| | - Yan-Xia Liu
- Department of General Medical, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Bin Yuan
- Department of Neurology II, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Henan Key Laboratory of Neural Regeneration, Xinxiang, China
| | - Jian-Hua Zhao
- Department of Neurology II, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Henan Key Laboratory of Neural Regeneration, Xinxiang, China.
| |
Collapse
|
104
|
Evzelman MA, Mityaeva EV, Lashkhiia IB, Kamchatnov PR. Acute cerebral ischemia and inflammation. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:73-80. [DOI: 10.17116/jnevro201911912273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
105
|
Ren Z, Chen L, Wang Y, Wei X, Zeng S, Zheng Y, Gao C, Liu H. Activation of the Omega-3 Fatty Acid Receptor GPR120 Protects against Focal Cerebral Ischemic Injury by Preventing Inflammation and Apoptosis in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 202:747-759. [PMID: 30598514 DOI: 10.4049/jimmunol.1800637] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/22/2018] [Indexed: 12/28/2022]
Abstract
G protein-coupled receptor 120 (GPR120) has been shown to negatively regulate inflammation and apoptosis, but its role in cerebral ischemic injury remains unclear. Using an in vivo model of middle cerebral artery occlusion (MCAO) and an in vitro model of oxygen-glucose deprivation (OGD), we investigated the potential role and molecular mechanisms of GPR120 in focal cerebral ischemic injury. Increased GPR120 expression was observed in microglia and neurons following MCAO-induced ischemia in wild type C57BL/6 mice. Treatment with docosahexaenoic acid (DHA) inhibited OGD-induced inflammatory response in primary microglia and murine microglial BV2 cells, whereas silencing of GPR120 strongly exacerbated the inflammation induced by OGD and abolished the anti-inflammatory effects of DHA. Mechanistically, DHA inhibited OGD-induced inflammation through GPR120 interacting with β-arrestin2. In addition to its anti-inflammatory function, GPR120 also played a role in apoptosis as its knockdown impaired the antiapoptotic effect of DHA in OGD-induced rat pheochromocytoma (PC12) cells. Finally, using MCAO mouse model, we demonstrated that GPR120 activation protected against focal cerebral ischemic injury by preventing inflammation and apoptosis. Our study indicated that pharmacological targeting of GPR120 may provide a novel approach for the treatment of patients with ischemic stroke.
Collapse
Affiliation(s)
- Zhiping Ren
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Lin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yimeng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xinbing Wei
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Shenglan Zeng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yi Zheng
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Chengjiang Gao
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; .,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China;
| |
Collapse
|
106
|
Sun M, Shinoda Y, Fukunaga K. KY-226 Protects Blood-brain Barrier Function Through the Akt/FoxO1 Signaling Pathway in Brain Ischemia. Neuroscience 2018; 399:89-102. [PMID: 30579831 DOI: 10.1016/j.neuroscience.2018.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/12/2018] [Accepted: 12/16/2018] [Indexed: 01/05/2023]
Abstract
KY-226 is a protein tyrosine phosphatase 1B (PTP1B) inhibitor that protects neurons from cerebral ischemic injury. KY-226 restores Akt (protein kinase B) phosphorylation and extracellular signal-regulated kinase (ERK) reduction in transient middle cerebral artery occlusion (tMCAO) damage. However, the mechanisms underlying the neuroprotective effects of KY-226 are unclear. To address this, the effects of KY-226 on blood-brain barrier (BBB) dysfunction were examined in tMCAO mice. KY-226 (10 mg/kg, i.p.) was administered to ICR mice 30 min after 2 h of tMCAO. To assess Akt or ERK involvement, wortmannin (i.c.v.) or U0126 (i.v.), selective inhibitors of PI3K and ERK, respectively, were administered to mice 30 min before ischemia. BBB integrity was assessed by Evans blue leakage 24 h post-reperfusion. The levels of tight junction (TJ) proteins, ZO-1 and occludin, were measured by western blotting; ZO-1 mRNA level was measured by RT-PCR. Compared to vehicle, KY-226 treatment prevented BBB breakdown and reduction in TJ protein levels. KY-226 treatment restored ZO-1 mRNA levels post-reperfusion. Pre-administration of wortmannin or U0126 blocked the protective effects of KY-226 on ZO-1 protein and mRNA reduction in tMCAO mice. In bEnd.3 cells, lipopolysaccharide treatment reduced mRNA and protein levels of ZO-1, an effect rescued by KY-226 treatment. Further, KY-226 treatment restored phosphorylation of pAkt (T308) and its downstream target forkhead box protein O1 (FoxO1) (S256) in bEnd.3 cells. Collectively, we demonstrate that KY-226 protects BBB integrity by restoration of TJ proteins, an effect partly mediated by Akt/FoxO1 pathway activation. Thus, protection of BBB integrity likely underlies KY-226-induced neuroprotection in tMCAO mice.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan.
| |
Collapse
|
107
|
Schultz NEØ, Hasseldam H, Rasmussen RS, Vindegaard N, McWilliam O, Iversen HK, Johansen FF. Statin treatment before stroke reduces pro-inflammatory cytokine levels after stroke. Neurol Res 2018; 41:289-297. [DOI: 10.1080/01616412.2018.1558000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
| | - Henrik Hasseldam
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Nina Vindegaard
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Oskar McWilliam
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
108
|
Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol 2018; 316:C135-C153. [PMID: 30379577 DOI: 10.1152/ajpcell.00136.2018] [Citation(s) in RCA: 481] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
As part of the neurovascular unit, the blood-brain barrier (BBB) is a unique, dynamic regulatory boundary that limits and regulates the exchange of molecules, ions, and cells between the blood and the central nervous system. Disruption of the BBB plays an important role in the development of neurological dysfunction in ischemic stroke. Blood-borne substances and cells have restricted access to the brain due to the presence of tight junctions between the endothelial cells of the BBB. Following stroke, there is loss of BBB tight junction integrity, leading to increased paracellular permeability, which results in vasogenic edema, hemorrhagic transformation, and increased mortality. Thus, understanding principal mediators and molecular mechanisms involved in BBB disruption is critical for the development of novel therapeutics to treat ischemic stroke. This review discusses the current knowledge of how neuroinflammation contributes to BBB damage in ischemic stroke. Specifically, we provide an updated overview of the role of cytokines, chemokines, oxidative and nitrosative stress, adhesion molecules, matrix metalloproteinases, and vascular endothelial growth factor as well as the role of different cell types in the regulation of BBB permeability in ischemic stroke.
Collapse
Affiliation(s)
- Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Sylvain Doré
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida.,Departments of Anesthesiology, Neurology, Psychiatry, Psychology, and Pharmaceutics, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| |
Collapse
|
109
|
Cheon SY, Kim EJ, Kim SY, Kim JM, Kam EH, Park JK, Koo BN. Apoptosis Signal-regulating Kinase 1 Silencing on Astroglial Inflammasomes in an Experimental Model of Ischemic Stroke. Neuroscience 2018; 390:218-230. [PMID: 30172704 DOI: 10.1016/j.neuroscience.2018.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/14/2018] [Accepted: 08/21/2018] [Indexed: 12/21/2022]
Abstract
Activation of the inflammasome complex contributes to the inflammatory response and cell death under pathologic conditions. The nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 2 (NLRP2) inflammasome is activated in astrocytes after cerebral ischemia, which can aggravate ischemic damage. Apoptosis signal-regulating kinase 1 (ASK1) is an early activator and immune-regulator after ischemic injury, that can lead to cell death. The objective of the present study was to evaluate the role of ASK1 in controlling NLRP2 inflammasomes in astrocytes after cerebral ischemia. In a mouse model of ischemic stroke, the levels of NLRP2 inflammasome components, and interleukin (IL)-1β and IL-18, were quantified in different brain regions. In addition, an astrocyte cell line was subjected to oxygen-glucose deprivation and reperfusion (OGD/R) injury, and the levels of NLRP2 inflammasome factors, IL-1β and IL-18 were evaluated. Ischemic brain injury activated astrocytes. The levels of NLRP2 inflammasome components, IL-1β and IL-18 productions, and cell death increased in the cortex and striatum after ischemic injury. In cultured astrocytes, NLRP2 inflammasome components, IL-1β and IL-18 levels were upregulated after OGD/R. ASK1 silencing or inhibition efficiently reduced NLRP2 inflammasome components and pro-inflammatory cytokine levels in mice and cultured astrocytes. Our findings identify a key role for ASK1 in regulating astroglial inflammasomes after cerebral ischemia. We suggest ASK1 as one of the main targets for astroglial inflammasomes in ischemic stroke.
Collapse
Affiliation(s)
- So Yeong Cheon
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Min Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Kam
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong-Kwang Park
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bon-Nyeo Koo
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
110
|
Zhang YY, Huang NN, Fan YC, Li YS, Zhao J, Wang D, Zhang F, Li XH. Peripheral tumor necrosis factor-a-induced protein 8-like 2 mRNA level for predicting 3-month mortality of patients with acute ischemic stroke. J Neurol 2018; 265:2573-2586. [PMID: 30171409 DOI: 10.1007/s00415-018-9036-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 12/31/2022]
|
111
|
Li SJ, Zhang YF, Ma SH, Yi Y, Yu HY, Pei L, Feng D. The role of NLRP3 inflammasome in stroke and central poststroke pain. Medicine (Baltimore) 2018; 97:e11861. [PMID: 30113480 PMCID: PMC6112889 DOI: 10.1097/md.0000000000011861] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND NLRP3 inflammasome plays a prominent role in the pathogenesis and progression of many diseases, such as type 2 diabetes mellitus, obesity, atherosclerosis, and Alzheimer's disease. However, little knowledge is known about the role of NLRP3 inflammasome in central post-stroke pain (CPSP). METHODS We selected relevant studies by searching PubMed, Embase, and Medline from inception through February, 2018. We systematically reviewed available publications according to the terms "NLRP3 inflammasome" and "stroke" or "central post-stroke pain" in the title/abstract field. RESULTS We reviewed the articles and put forward two possible ways for NLRP3 inflammasome in CPSP. One way is that NLRP3 activation causes cerebral cortex injure, decreasing descending projection fiber to thalamus. Such condition may let GABAergic releases reduce, making the ventral basal (VB) neurons excitability increased. Finally, CPSP occur. Another way is that NLRP3 inflammasome leads to thalamic lesion and strengthens inflammatory response of microglia at the same time. Persistent inflammation causes GABAergic alteration in thalamus reticular neurons (TRN) to restrain VB interneurons functions, contributing to CPSP. CONCLUSIONS These possible mechanisms will help become knowledgeable about the occurrence CPSP and provide potential therapy for CPSP.
Collapse
Affiliation(s)
- Shao-jun Li
- Department of Pain Management, Wuhan First Hospital
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-fen Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Se-hui Ma
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Yi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-yan Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Pei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Feng
- Department of Pain Management, Wuhan First Hospital
- The Institute for Brain Research (IBR), Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
112
|
Swanton T, Cook J, Beswick JA, Freeman S, Lawrence CB, Brough D. Is Targeting the Inflammasome a Way Forward for Neuroscience Drug Discovery? SLAS DISCOVERY 2018; 23:991-1017. [PMID: 29969573 DOI: 10.1177/2472555218786210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuroinflammation is becoming increasingly recognized as a critical factor in the pathology of both acute and chronic neurological conditions. Inflammasomes such as the one formed by NACHT, LRR, and PYD domains containing protein 3 (NLRP3) are key regulators of inflammation due to their ability to induce the processing and secretion of interleukin 1β (IL-1β). IL-1β has previously been identified as a potential therapeutic target in a variety of conditions due to its ability to promote neuronal damage under conditions of injury. Thus, inflammasome inhibition has the potential to curtail inflammatory signaling, which could prove beneficial in certain diseases. In this review, we discuss the evidence for inflammasome contributions to the pathology of neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease, epilepsy, and acute degeneration following brain trauma or stroke. In addition, we review the current landscape of drug development targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Tessa Swanton
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James Cook
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James A Beswick
- 2 Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sally Freeman
- 2 Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Catherine B Lawrence
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - David Brough
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
113
|
Lü C, Liu Q, Zeng X. [Effects of interleukin 10 gene modified bone marrow mesenchymal stem cells on expression of inflammatory cytokines and neuronal apoptosis in rats after cerebral ischemia reperfusion injury]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 31:240-245. [PMID: 29786261 DOI: 10.7507/1002-1892.201605095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To explore the effects of interleukin 10 (IL-10) gene modified bone marrow mesenchymal stem cells (BMSCs) on the expression of inflammatory cytokines and neuronal apoptosis in rats after cerebral ischemia reperfusion injury. Methods BMSCs were cultured by whole bone marrow adherence screening method. The properties of BMSCs were identified by immunocytochemical methods. BMSCs at passage 3 were transfected with recombinant adenovirus IL-10 gene (AdIL-10-BMSCs). The model of middle cerebral artery occlusion was made in 40 adult male Sprague Dawley rats by thread embolism method. The rats were randomly divided into 4 groups ( n=10). At 3 hours after modelling, the rats of groups A, B, C, and D received tail intravenous injection of 1 mL L-DMEM medium containing 10% FBS, 61.78 ng IL-10, 1 mL BMSCs suspension (2×10 6 cells/mL), and 1 mL AdIL-10-BMSCs cell suspension (2×10 6 cells/mL), respectively. The cells were labelled with BrdU before cell transplantation in groups C and D. At 7 days after reperfusion, the brain tissue was harvested to detect the expression of OX42 by immunohistochemical assay, to determine the concentration of tumor necrosis factor α (TNF-α) and IL-1β by ELISA, and to detect the apoptosis by TUNEL assay. BrdU labelled cells were observed by immunofluorescence staining in groups C and D. Results BrdU labelled positive cells with green fluorescence were observed in the brain tissue of groups C and D, which mainly distributed in the striatum, cerebral cortex, and subcortex around the infarction area. The number of OX42 positive cells was significantly less in groups B, C, and D than group A ( P<0.05), and in group D than groups B and C ( P<0.05). Compared with the other 3 groups, the contents of TNF-α and IL-1β significantly decreased in group D ( P<0.05). TUNEL assay showed that the apoptotic cells (TUNEL positive cells) were mainly seen in the striatum and fronto parietal subcortical tissues (equivalent to ischemic penumbra). The number of TUNEL positive cells in group D was significantly less than that in groups A, B, and C ( P<0.05). Conclusion AdIL-10-BMSCs can inhibit secretion of TNF-α and IL-1β from microglial cells and inhibit the nerve cell apoptosis around infarct brain tissue, which might contribute to its protective role upon cerebral ischemia reperfusion injury.
Collapse
Affiliation(s)
- Cui Lü
- Electrocardiographic Room, Affiliated Hospital of Weifang Medical University, Weifang Shandong, 261031, P.R.China
| | - Qian Liu
- Electrocardiographic Room, Affiliated Hospital of Weifang Medical University, Weifang Shandong, 261031, P.R.China
| | - Xianwei Zeng
- Electrocardiographic Room, Affiliated Hospital of Weifang Medical University, Weifang Shandong, 261031,
| |
Collapse
|
114
|
Xu W, Gao L, Zheng J, Li T, Shao A, Reis C, Chen S, Zhang J. The Roles of MicroRNAs in Stroke: Possible Therapeutic Targets. Cell Transplant 2018; 27:1778-1788. [PMID: 29871520 PMCID: PMC6300776 DOI: 10.1177/0963689718773361] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Stroke is one of the most devastating diseases worldwide. In recent years, a great number of studies have focused on the effects of microRNAs (miRNAs) on stroke and the results demonstrated that the expressions of miRNAs are associated with the prognosis of stroke. In the present study, we review relevant articles regarding miRNAs and stroke and will explain the complex link between both. The miRNAs participate extensively in the pathophysiology following the stroke, including apoptosis, neuroinflammation, oxidative stress, blood–brain barrier (BBB) disruption and brain edema. The information about the stroke–miRNA system may be helpful for therapeutic and diagnostic methods in stroke treatment.
Collapse
Affiliation(s)
- Weilin Xu
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liansheng Gao
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingwei Zheng
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tao Li
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cesar Reis
- 4 Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Sheng Chen
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- 1 Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,2 Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.,3 Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
115
|
Karlsson L, González-Alvarado MN, Larrosa-Flor M, Osman A, Börjesson M, Blomgren K, Kuhn HG. Constitutive PGC-1α Overexpression in Skeletal Muscle Does Not Improve Morphological Outcome in Mouse Models of Brain Irradiation or Cortical Stroke. Neuroscience 2018; 384:314-328. [PMID: 29859976 DOI: 10.1016/j.neuroscience.2018.05.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/19/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022]
Abstract
Physical exercise can improve morphological outcomes after ischemic stroke and ameliorate irradiation-induced reduction of hippocampal neurogenesis in rodents, but the mechanisms underlying these effects remain largely unknown. The transcription factor peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) is considered to be one of the central factors responsible for exercise-induced benefits in skeletal muscle, including the release of neurotrophic factors into the circulation. In order to test if PGC-1α overexpression in skeletal muscle could simulate the exercise-induced effects on recovery after cranial irradiation and stroke, we used male adult transgenic mice overexpressing murine PGC-1α under the control of muscle creatinine kinase promoter and subjected them to either whole brain irradiation at a dose of 4 Gy or photothrombotic stroke to the sensory motor cortex. Muscular PGC-1α overexpression did not ameliorate irradiation-induced reduction of newborn BrdU-labeled cells in the dentate gyrus, immature neurons, or newborn mature neurons. In the stroke model, muscular overexpression of PGC-1α resulted in an increased infarct size without any changes in microglia activation or reactive astrocytosis. No difference could be detected in the number of migrating neural progenitor cells from the subventricular zone to the lesioned neocortex or in vascular density of the contralateral neocortex in comparison to wildtype animals. We conclude that forced muscular overexpression of PGC-1α does not have a beneficial effect on hippocampal neurogenesis after irradiation, but rather a detrimental effect on the infarct volume after stroke in mice. This suggests that artificial muscle activation through the PGC-1α pathway is not sufficient to mimic exercise-induced recovery after cranial irradiation and stroke.
Collapse
Affiliation(s)
- Lars Karlsson
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden; The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, 416 85 Gothenburg, Sweden.
| | | | - Mar Larrosa-Flor
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden
| | - Ahmed Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Mats Börjesson
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden; Center for Health and Performance, Department of Food and Nutrition, University of Gothenburg, Box 300, 405 30 Gothenburg, Sweden; Sahlgrenska University Hospital/Östra, 416 50 Gothenburg, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Hans Georg Kuhn
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden
| |
Collapse
|
116
|
Han J, Yin QH, Fang Y, Shou WQ, Zhang CC, Guo FQ. Atorvastatin protects BV‑2 mouse microglia and hippocampal neurons against oxygen‑glucose deprivation‑induced neuronal inflammatory injury by suppressing the TLR4/TRAF6/NF‑κB pathway. Mol Med Rep 2018; 18:1058-1066. [PMID: 29845194 DOI: 10.3892/mmr.2018.9055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/08/2018] [Indexed: 11/06/2022] Open
Abstract
Atorvastatin is a member of the statin class of drugs, which competitively inhibit the activity of 5‑hydroxy‑3‑methylglutaryl‑coenzyme A reductase. The aim of the present study was to assess whether atorvastatin may protect BV‑2 microglia and hippocampal neurons against oxygen‑glucose deprivation (OGD)‑induced neuronal inflammatory injury and to determine the underlying mechanisms by which its effects are produced. Cell viability and apoptotic ability were assessed using an MTT assay and annexin V‑fluorescein isothiocyanate/propidium iodide double staining followed by flow cytometry, respectively. The expression of inflammation and apoptosis‑associated mRNAs and proteins were assessed using reverse transcription‑quantitative polymerase chain reaction and western blotting, and the expression of inflammatory factors was determined using ELISA. The results of the current study revealed that atorvastatin treatment suppressed the viability of OGD BV‑2 microglia and hippocampal neurons. Furthermore, atorvastatin treatment reduced the expression of proinflammatory factors in OGD BV‑2 microglia. Additionally, it was demonstrated to downregulate the toll‑like receptor 4 (TLR4)/tumor necrosis factor receptor‑associated factor 6 (TRAF6)/nuclear factor‑κB (NF‑κB) pathway in OGD BV‑2 microglia. Atorvastatin also inhibited the apoptosis of OGD hippocampal neurons by regulating the expression of apoptosis‑associated proteins. It was concluded that atorvastatin treatment may protect BV‑2 microglia and hippocampal neurons from OGD‑induced neuronal inflammatory injury by suppressing the TLR4/TRAF6/NF‑κB pathway. This may provide a potential strategy for the treatment of neuronal injury.
Collapse
Affiliation(s)
- Jian Han
- Department of Neurology, Shaoxing Municipal Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Qi-Hua Yin
- Department of Neurology, Shaoxing Municipal Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yang Fang
- Department of Neurology, Shaoxing Municipal Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Wei-Qing Shou
- Department of Neurology, Shaoxing Municipal Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Cong-Cong Zhang
- Department of Neurology, Shaoxing Municipal Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Fu-Qiang Guo
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
117
|
Ding Y, Qian J, Li H, Shen H, Li X, Kong Y, Xu Z, Chen G. Effects of SC99 on cerebral ischemia-perfusion injury in rats: Selective modulation of microglia polarization to M2 phenotype via inhibiting JAK2-STAT3 pathway. Neurosci Res 2018; 142:58-68. [PMID: 29763638 DOI: 10.1016/j.neures.2018.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/29/2022]
Abstract
Inhibition of Janus kinases 2-Signal transducers and activators of transcription3 (JAK2-STAT3) pathway has been shown to exert anti-inflammatory actions. SC99, a novel specific inhibitor targeting JAK2-STAT3 pathway, has been verified to negatively modulate platelet activation and aggregation in vitro. In current study, a middle cerebral artery occlusion and reperfusion (MCAO/R) model was established in Sprague Dawley rats and primary cultured microglia was exposed to oxygen and glucose deprivation (OGD/R) in vitro. Different dosages were employed to detect the effects of SC99 on cerebral ischemia-perfusion (I/R) injury and evaluate the underlying mechanisms. Our results showed that intracerebroventricular injection of SC99 (10 mmol/L, 15 μL) produced an effective inhibitory effect on the phosphorylation of JAK2 and STAT3. Correspondingly, SC99 ameliorated neuronal apoptosis and degeneration, neurobehavioral deficits, inflammatory response and brain edema. And SC99 promoted microglia polarization to an anti-inflammatory M2 phenotype. We concluded that SC99 could alleviate brain damage and play an anti-inflammatory action by promoting microglia polarization to an anti-inflammatory phenotype after I/R injury, which provides an emerging and promising alternative to protect the brain against MCAO/R injury in the future investigations.
Collapse
Affiliation(s)
- Yiping Ding
- Department of Neurology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Jinhong Qian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Yan Kong
- Department of Neurology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Zhuan Xu
- Department of Neurology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| |
Collapse
|
118
|
Spaide RF, Fujimoto JG, Waheed NK, Sadda SR, Staurenghi G. Optical coherence tomography angiography. Prog Retin Eye Res 2018; 64:1-55. [PMID: 29229445 PMCID: PMC6404988 DOI: 10.1016/j.preteyeres.2017.11.003] [Citation(s) in RCA: 1014] [Impact Index Per Article: 169.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 02/07/2023]
Abstract
Optical coherence tomography (OCT) was one of the biggest advances in ophthalmic imaging. Building on that platform, OCT angiography (OCTA) provides depth resolved images of blood flow in the retina and choroid with levels of detail far exceeding that obtained with older forms of imaging. This new modality is challenging because of the need for new equipment and processing techniques, current limitations of imaging capability, and rapid advancements in both imaging and in our understanding of the imaging and applicable pathophysiology of the retina and choroid. These factors lead to a steep learning curve, even for those with a working understanding dye-based ocular angiography. All for a method of imaging that is a little more than 10 years old. This review begins with a historical account of the development of OCTA, and the methods used in OCTA, including signal processing, image generation, and display techniques. This forms the basis to understand what OCTA images show as well as how image artifacts arise. The anatomy and imaging of specific vascular layers of the eye are reviewed. The integration of OCTA in multimodal imaging in the evaluation of retinal vascular occlusive diseases, diabetic retinopathy, uveitis, inherited diseases, age-related macular degeneration, and disorders of the optic nerve is presented. OCTA is an exciting, disruptive technology. Its use is rapidly expanding in clinical practice as well as for research into the pathophysiology of diseases of the posterior pole.
Collapse
Affiliation(s)
- Richard F Spaide
- Vitreous, Retina, Macula Consultants of New York, New York, NY, United States.
| | - James G Fujimoto
- Department of Electrical Engineering & Computer Science and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge MA, United States
| | - Nadia K Waheed
- The Department of Ophthalmology, Tufts University School of Medicine, Boston MA, United States
| | - Srinivas R Sadda
- Doheny Eye Institute, University of California - Los Angeles, Los Angeles, CA, United States
| | - Giovanni Staurenghi
- Eye Clinic, Department of Biomedical and Clinical Sciences "Luigi Sacco", Luigi Sacco Hospital, University of Milan, Milan, Italy
| |
Collapse
|
119
|
Osier N, Motamedi V, Edwards K, Puccio A, Diaz-Arrastia R, Kenney K, Gill J. Exosomes in Acquired Neurological Disorders: New Insights into Pathophysiology and Treatment. Mol Neurobiol 2018; 55:9280-9293. [PMID: 29663285 DOI: 10.1007/s12035-018-1054-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 03/29/2018] [Indexed: 01/08/2023]
Abstract
Exosomes are endogenous nanovesicles that play critical roles in intercellular signaling by conveying functional genetic information and proteins between cells. Exosomes readily cross the blood-brain barrier and have promise as therapeutic delivery vehicles that have the potential to specifically deliver molecules to the central nervous system (CNS). This unique feature also makes exosomes attractive as biomarkers in diagnostics, prognostics, and therapeutics in the context of multiple significant public health conditions, including acquired neurological disorders. The purpose of this review is to summarize the state of the science surrounding the relevance of extracellular vesicles (EVs), particularly exosomes, to acquire neurological disorders, specifically traumatic brain injury (TBI), spinal cord injury (SCI), and ischemic stroke. In total, ten research articles were identified that examined exosomes in the context of TBI, SCI, or stroke; these manuscripts were reviewed and synthesized to further understand the current role of exosomes in the context of acquired neurological disorders. Of the ten published studies, four focused exclusively on TBI, one on both TBI and SCI, and five on ischemic stroke; notably, eight of the ten studies were limited to pre-clinical samples. The present review is the first to discuss the current body of knowledge surrounding the role of exosomes in the pathophysiology, diagnosis, and prognosis, as well as promising therapeutic strategies in TBI, SCI, and stroke research.
Collapse
Affiliation(s)
- Nicole Osier
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA. .,University of Texas at Austin, Austin, TX, USA.
| | - Vida Motamedi
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA
| | - Katie Edwards
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA.,Healthcare Genetics Doctoral Program, Clemson University School of Nursing, 508 Edwards, Clemson, SC, 29631, USA
| | - Ava Puccio
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite B-400, Pittsburgh, PA, 15213, USA
| | - Ramon Diaz-Arrastia
- University of Pennsylvania School of Medicine, Suite 205 Medical Office Building, 51 N 39TH ST, Philadelphia, PA, 19104, USA
| | - Kimbra Kenney
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Building 51, Room 2306, 4860 South Palmer Road, Bethesda, MD, 20889-5649, USA
| | - Jessica Gill
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA
| |
Collapse
|
120
|
Gupta S, Upadhayay D, Sharma U, Jagannathan NR, Gupta YK. Citalopram attenuated neurobehavioral, biochemical, and metabolic alterations in transient middle cerebral artery occlusion model of stroke in male Wistar rats. J Neurosci Res 2018; 96:1277-1293. [PMID: 29656429 DOI: 10.1002/jnr.24226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/15/2017] [Accepted: 01/30/2018] [Indexed: 11/08/2022]
Abstract
Oxidative stress and inflammation are implicated as cardinal mechanisms of neuronal death following stroke. In the present study citalopram (Cit) was investigated in a 2 h middle cerebral artery occlusion (MCAo) model of stroke in male Wistar rats. Pretreatment, posttreatment (Post Cit) and pre plus posttreatment (Pre + Post Cit) with Cit were evaluated for its neuroprotective effect. In pretreatment protocol, effect of Cit at three doses (2, 4, and 8 mg/kg) administered i.p., 1 h prior to MCAo was evaluated using neurological deficit score (NDS), motor deficit paradigms, and cerebral infarction 24 h post-MCAo. In posttreatment and pre plus posttreatment protocol, the effective dose of Cit (4 mg/kg) was administered i.p., 0.5 h post-reperfusion (Post Cit) only, and 1 h prior to MCAo and again at 0.5 h post-reperfusion (Pre + Post Cit), respectively. These two groups were assessed for NDS and cerebral infarction. Though NDS was significantly reduced in both Post Cit and Pre + Post Cit groups, significant reduction in cerebral infarction was evident only in Pre + Post Cit group. Infarct volume assessed by magnetic resonance imaging was significantly attenuated in Pre + Post Cit group (10.6 ± 1.1%) compared to MCAo control group (18.5 ± 3.0%). Further, Pre + Post Cit treatment significantly altered 17 metabolites along with attenuation of malondialdehyde, reduced glutathione, matrix metalloproteinases, and apoptotic markers as compared to MCAo control. These results support the neuroprotective effect of Cit, mediated through amelioration of oxidative stress, inflammation, apoptosis, and altered metabolic profile.
Collapse
Affiliation(s)
- Sangeetha Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Deepti Upadhayay
- Department of NMR & MRI Facility, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Uma Sharma
- Department of NMR & MRI Facility, All India Institute of Medical Sciences, New Delhi-110029, India
| | | | - Yogendra Kumar Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
121
|
Lu X, Gu R, Hu W, Sun Z, Wang G, Wang L, Xu Y. Upregulation of heme oxygenase-1 protected against brain damage induced by transient cerebral ischemia-reperfusion injury in rats. Exp Ther Med 2018; 15:4629-4636. [PMID: 29805479 PMCID: PMC5958683 DOI: 10.3892/etm.2018.6049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 07/07/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to identify the effect of heme oxygenase (HO)-1 gene on cerebral ischemia-reperfusion injury. Sprague-Dawley rats were divided randomly into four groups: Sham group, vehicle group, empty adenovirus vector (Ad) group and recombinant HO-1 adenovirus (Ad-HO-1) transfection group. Rats in the vehicle, Ad and Ad-HO-1 groups were respectively injected with saline, Ad or Ad-HO-1 for 3 days prior to cerebral ischemia-reperfusion injury. Subsequently, the middle cerebral artery occlusion method was used to establish the model of cerebral ischemia-reperfusion injury. Following the assessment of neurological function, rats were sacrificed, and the infarction volume and apoptotic index in rat brains were measured. Furthermore, the protein expression levels of HO-1 in brain tissues were detected using western blot analysis. Results indicated that the neurological score of the Ad-HO-1 group was significantly increased compared with the Ad or vehicle groups, respectively (P<0.001). The volume of cerebral infarction and the index score of neuronal apoptosis in the vehicle and Ad groups was significantly increased compared with the Ad-HO-1 group (P<0.01). The death of neuronal cells following cerebral ischemia-reperfusion injury reduced remarkably induced by over-expression of HO-1. These findings suggest a neuroprotective role of HO-1 against brain injury induced by transient cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xiufang Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Renjun Gu
- Department of Neurology, The Second Affiliated Hospital of Xinxiang Medical College, Xinxiang, Henan 453100, P.R. China
| | - Weimin Hu
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zhitang Sun
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Gaiqing Wang
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Li Wang
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
122
|
Cell Type-Specific Mechanisms in the Pathogenesis of Ischemic Stroke: The Role of Apoptosis Signal-Regulating Kinase 1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2596043. [PMID: 29743976 PMCID: PMC5883936 DOI: 10.1155/2018/2596043] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 02/10/2018] [Accepted: 02/22/2018] [Indexed: 12/19/2022]
Abstract
Stroke has become a more common disease worldwide. Despite great efforts to develop treatment, little is known about ischemic stroke. Cerebral ischemia activates multiple cascades of cell type-specific pathomechanisms. Ischemic brain injury consists of a complex series of cellular reactions in various cell types within the central nervous system (CNS) including platelets, endothelial cells, astrocytes, neutrophils, microglia/macrophages, and neurons. Diverse cellular changes after ischemic injury are likely to induce cell death and tissue damage in the brain. Since cells in the brain exhibit different functional roles at distinct time points after injury (acute/subacute/chronic phases), it is difficult to pinpoint genuine roles of cell types after brain injury. Many experimental studies have shown the association of apoptosis signal-regulating kinase 1 (ASK1) with cellular pathomechanisms after cerebral ischemia. Blockade of ASK1, by either pharmacological or genetic manipulation, leads to reduced ischemic brain injury and subsequent neuroprotective effects. In this review, we present the cell type-specific pathophysiology of the early phase of ischemic stroke, the role of ASK1 suggested by preclinical studies, and the potential use of ASK suppression, either by pharmacologic or genetic suppression, as a promising therapeutic option for ischemic stroke recovery.
Collapse
|
123
|
Xu H, Qin W, Hu X, Mu S, Zhu J, Lu W, Luo Y. Lentivirus-mediated overexpression of OTULIN ameliorates microglia activation and neuroinflammation by depressing the activation of the NF-κB signaling pathway in cerebral ischemia/reperfusion rats. J Neuroinflammation 2018; 15:83. [PMID: 29544517 PMCID: PMC5856386 DOI: 10.1186/s12974-018-1117-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/06/2018] [Indexed: 01/05/2023] Open
Abstract
Background Ischemic stroke-induced neuroinflammation is mainly mediated by microglial cells. The nuclear factor kappa B (NF-κB) pathway is the key transcriptional pathway that initiates inflammatory responses following cerebral ischemia. OTULIN, a critical negative regulator of the NF-κΒ signaling pathway, exerts robust effects on peripheral immune cell-mediated inflammation and is regarded as an essential mediator for repressing inflammation in vivo. The effect of OTULIN on inflammatory responses in the central nervous system (CNS) was previously unstudied. This current study investigated the anti-inflammatory effect of OTULIN both in vitro and in vivo in ischemic stroke models. Methods Sprague-Dawley (SD) rats were subjected to transient middle cerebral artery occlusion (tMCAO) or an intraperitoneal injection of lipopolysaccharide (LPS). Overexpression of the OTULIN gene was utilized to observe the effect of OTULIN on ischemic stroke outcomes. The effect of OTULIN overexpression on microglia-mediated neuroinflammation was examined in rat primary microglia (PM) and in the microglial cell line N9 after induction by oxygen-glucose deprivation (OGD)-treated neuronal medium. The activation and inflammatory responses of microglia were detected using immunofluorescence, ELISA, and qRT-PCR. The details of molecular mechanism were assessed using Western blotting. Results In the tMCAO rats, the focal cerebral ischemia/reperfusion injury induced a continuous increase in OTULIN expression within 72 h, and OTULIN expression was increased in activated microglial cells. OTULIN overexpression obviously decreased the cerebral infarct volume, improved the neurological function deficits, and reduced neuronal loss at 72 h after reperfusion, and it also inhibited the activation of microglia and attenuated the release of TNF-α, IL-1β, and IL-6 by suppressing the NF-κB pathway at 24 h after tMCAO. In vitro, OTULIN overexpression inhibited the microglia-mediated neuroinflammation by reducing the production of TNF-α, IL-1β, and IL-6 via depressing the NF-κB pathway in both PM and N9 cells. Conclusions OTULIN provides a potential therapeutic target for ischemic brain injury by ameliorating the excessive activation of microglial cells and neuroinflammation through repressing the NF-κB signaling pathway. Electronic supplementary material The online version of this article (10.1186/s12974-018-1117-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongbei Xu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenyi Qin
- Department of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiao Hu
- Department of Neurology, Guizhou Provincial People's hospital, Guizhou, 50002, China
| | - Song Mu
- Department of Anus & Intestine surgery, the Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, China
| | - Jun Zhu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenhao Lu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong Luo
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
124
|
MICROVASCULAR FLOW ABNORMALITIES ASSOCIATED WITH RETINAL VASCULITIS: A Potential of Mechanism of Retinal Injury. Retina 2018; 37:1034-1042. [PMID: 27759582 DOI: 10.1097/iae.0000000000001359] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To investigate the structural optical coherence tomography and related microvascular flow characteristics in eyes with retinal vasculitis. METHODS Regions involved with perivascular infiltration in eyes with retinal vasculitis, but no evidence of large vessel occlusion were evaluated with optical coherence tomography (OCT), OCT angiography, and fluorescein angiography. RESULTS Ten eyes of 5 patients with a variety of etiologies of retinal vasculitis were evaluated. These patients did not have either cotton wool spots or deeper placoid areas of retinal ischemia. Around large vessels there was perivascular infiltration with leakage and staining seen during fluorescein angiography. Structural OCT showed slight thickening with loss of visualization of normal retinal laminations. OCT angiography showed a lack of flow signal in capillary sized vessels in the same areas. Treatment resulted in a rapid thinning of the affected areas, with the inner and middle layers of the retina becoming thinner than surrounding uninvolved areas. OCT angiography did not show a return of capillary perfusion in these regions. The thickness change in the structural OCT as shown by a heat map had a pattern mimicking the original perivascular infiltration around large retinal vessels. CONCLUSION Capillary level perfusion abnormalities can develop in regions adjacent to large vessel inflammatory infiltrate that result in retinal thinning without the development of usual stigmata of acute microvascular flow obstruction such as cotton wool spots. This suggests that retinal damage may occur in retinal vasculitis that would not be recognized without using OCT-based imaging modalities.
Collapse
|
125
|
Neuroprotective effects of pretreatment with minocycline on memory impairment following cerebral ischemia in rats. Behav Pharmacol 2018; 28:214-222. [PMID: 28257293 DOI: 10.1097/fbp.0000000000000297] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cerebral ischemia leads to memory impairment that is associated with loss of hippocampal CA1 pyramidal neurons. Neuroinflammation and oxidative stress may be implicated in the pathogenesis of ischemia/reperfusion damage. Minocycline has anti-inflammatory and antioxidant properties. We investigated the neuroprotective effects of minocycline in rats subjected to cerebral ischemia/reperfusion injury. Thirty male rats were divided into three groups: control, sham, and minocycline-pretreated group. Minocycline (40 mg/kg) was injected intraperitoneally immediately before surgery, and then ischemia was induced by occlusion of common carotid arteries for 20 min. Seven days after reperfusion, the Morris water-maze task was used to evaluate memory. Nissl staining was also performed to analyze pyramidal cell damage. We measured the contents of malondialdehyde and proinflammatory cytokines in the hippocampus by the thiobarbituric acid method and enzyme-linked immunosorbent assay, respectively. Microglial activation was also investigated by Iba1 immunostaining. The results showed that pretreatment with minocycline prevented memory impairment induced by cerebral ischemia/reperfusion. Minocycline pretreatment also significantly attenuated ischemia-induced pyramidal cell death and microglial activation in the CA1 region and reduced the levels of malondialdehyde and proinflammatory cytokines (interleukin-1β and tumor necrosis factor-α) in the hippocampus of ischemic rats. Minocycline showed neuroprotective effects on cerebral ischemia-induced memory deficit probably through its anti-inflammatory and antioxidant activities.
Collapse
|
126
|
Xiong W, Wu Y, Xian W, Song L, Hu L, Pan S, Liu M, Yao S, Pei L, Shang Y. DAPK1-ERK signal mediates oxygen glucose deprivation reperfusion induced apoptosis in mouse N2a cells. J Neurol Sci 2018; 387:210-219. [PMID: 29571866 DOI: 10.1016/j.jns.2018.01.003] [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: 08/16/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 11/28/2022]
Abstract
AIMS Death-associated protein kinase 1 (DAPK1) is a kinase found to promote neuronal apoptosis induced by ischemia. Extracellular signal-regulated kinase (ERK) was identified as a key molecule in DAPK1 signaling. However, the mechanisms of neuronal ischemia reperfusion injury remain unknown. Here, we investigate the influence of DAPK1-ERK signal on neuronal apoptosis following ischemia reperfusion. METHODS Mouse N2a cells were used in this study and primary cultured neurons along with mice were adopted as supplements. Oxygen glucose deprivation (OGD) or administration of N-methyl-d-aspartate (NMDA) and glycine was performed on cells while middle cerebral artery occlusion (MCAO) model on mice. DAPK1 knocking down was achieved by lentiviral-delivered shRNA. Protein expressions were evaluated by western blots. Protein-protein binding was confirmed by co-immunoprecipitation and immunofluorescent assay. Apoptosis of cells was measured by flow cytometry and lacate dehydrogenase (LDH) leakage assay. RESULTS Ischemia reperfusion resulted in increased DAPK1 and ERK activation as well as aggravated apoptosis in a time-dependent manner. DAPK1 was proved to bind to ERK during reperfusion following OGD, MCAO and excitotoxicity model. Interception of this binding by knocking down DAPK1 led to nuclear translocation of ERK and reduced apoptosis. CONCLUSION Our study revealed the DAPK1-ERK signal as a potential mechanism contributing to neuronal apoptosis in response to ischemia reperfusion. Disruption of this signal pathway could be a promising therapeutic target against stroke.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjing Xian
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Limin Song
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lisha Hu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shangwen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Liu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanglong Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Pei
- Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
127
|
|
128
|
Peripheral immune cells infiltrate into sites of secondary neurodegeneration after ischemic stroke. Brain Behav Immun 2018; 67:299-307. [PMID: 28911981 DOI: 10.1016/j.bbi.2017.09.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 09/10/2017] [Accepted: 09/10/2017] [Indexed: 12/13/2022] Open
Abstract
Experimental stroke leads to microglia activation and progressive neuronal loss at sites of secondary neurodegeneration (SND). These lesions are remote from, but synaptically connected to, primary infarction sites. Previous studies have demonstrated that immune cells are present in sites of infarction in the first hours and days after stroke, and are associated with increased neurodegeneration in peri-infarct regions. However, it is not known whether immune cells are also present in more distal sites where SND occurs. Our study aimed to investigate whether immune cells are present in sites of SND and, if so, how these cell populations compare to those in the peri-infarct zone. Cells were isolated from the thalamus, the main site of SND, and remaining brain tissue 14days post-stroke. Analysis was performed using flow cytometry to quantify microglia, myeloid cell and lymphocyte numbers. We identified a substantial infiltration of immune cells in the ipsilateral (stroked) compared to the contralateral (control) thalamus, with a significant increase in the percentage of CD4+ and CD8+ T cells. This result was further quantified using immunofluorescent labelling of fixed tissue. In the remaining ipsilateral hemisphere tissue, there were significant increases in the frequency of CD4+ and CD8+ T lymphocytes, B lymphocytes, Ly6G+ neutrophils and both Ly6G-Ly6CLO and Ly6G-Ly6CHI monocytes. Our results indicate that infiltrating immune cells persist in ischemic tissue after the acute ischemic phase, and are increased in sites of SND. Importantly, immune cells have been shown to play pivotal roles in both damage and repair processes after stroke. Our findings indicate that immune cells may also be involved in the pathogenesis of SND and further clinical studies are warranted to characterise the nature of inflammatory cell infiltrates in human disease.
Collapse
|
129
|
Zhang YY, Huang NN, Zhao YX, Li YS, Wang D, Fan YC, Li XH. Elevated Tumor Necrosis Factor-a-induced Protein 8-like 2 mRNA from Peripheral Blood Mononuclear Cells in Patients with Acute Ischemic Stroke. Int J Med Sci 2018; 15:1713-1722. [PMID: 30588195 PMCID: PMC6299423 DOI: 10.7150/ijms.27817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022] Open
Abstract
Background: Tumor necrosis factor-a-induced protein 8-like 2 (TIPE2) is a novel regulator of immunity and protects against experimental stroke. However, the expression and function of TIPE2 in patients with acute ischemic stroke has not been well demonstrated. Methods: A total of 182 consecutive patients with acute ischemic stroke and 40 healthy controls were included during November 2015 to June 2016. The mRNA levels of TIPE2, interleukin(IL)-1β, IL-10, IL-6, nuclear factor(NF)-κβ, activator protein(AP)-1, interferon(IFN)-γ and tumor necrosis factor(TNF)-α from peripheral blood mononuclear cells were determined using real time quantitative reverse transcriptase polymerase chain reaction. The severity of stroke was assessed using the National Institutes of Health Stroke Scale (NIHSS) score. Results: The median mRNA levels of TIPE2, TNF-α, AP-1, IFN-γ and NF-κβ in patients with acute ischemic stroke were significantly higher than healthy controls (all P<0.001, respectively). Of note, TIPE2 mRNA showed an increasing trend on a time-dependent manner after the onset of stroke. Furthermore, TIPE2 mRNA was negatively associated with lesion volumes (r=-0.23, P<0.01), NIHSS(r=-0.15, P<0.05), TNF-α(r=-0.33,P<0.001), AP-1(r=-0.28,P<0.001), IFN-γ (r=-0.16, P<0.05) and NF-κβ (r=-0.13, P<0.05), but positively associated with IL-6(r=0.14, P<0.05) and IL-10(r=-0.31, P<0.001). Hierarchy cluster analysis showed that TIPE2 mRNA has nearest membership with TNF-α, followed by IL-6, NF-κβ, AP-1, IL-10, IL-1β and IFN-γ. In addition, TIPE2 mRNA in survivals (n=149) was significantly higher than nonsurvivals (n=33) (P<0.001), and showed a great odd ratio (0.52, 95% confidence interval: 0.349-0.760, P<0.001) on 3-month mortality. Conclusions: TIPE2 mRNA contributed to the immune response of stroke and might be a potential biomarker for the mortality of acute ischemic stroke.
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Na-Na Huang
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Yan-Xin Zhao
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Yan-Shuang Li
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Dong Wang
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiao-Hong Li
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| |
Collapse
|
130
|
Hwang SA, Kim CD, Lee WS. Caffeic acid phenethyl ester protects against photothrombotic cortical ischemic injury in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 22:101-110. [PMID: 29302217 PMCID: PMC5746507 DOI: 10.4196/kjpp.2018.22.1.101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 12/29/2022]
Abstract
In this study, we aimed to investigate the neuroprotective effects of caffeic acid phenethyl ester (CAPE), an active component of propolis purified from honeybee hives, on photothrombotic cortical ischemic injury in mice. Permanent focal ischemia was achieved in the medial frontal and somatosensory cortices of anesthetized male C57BL/6 mice by irradiation of the skull with cold light laser in combination with systemic administration of rose bengal. The animals were treated with CAPE (0.5–5 mg/kg, i.p.) twice 1 and 6 h after ischemic insult. CAPE significantly reduced the infarct size as well as the expression of tumor necrosis factor-α, hypoxiainducible factor-1α, monocyte chemoattractant protein-1, interleukin-1α, and indoleamine 2,3-dioxygenase in the cerebral cortex ipsilateral to the photothrombosis. Moreover, it induced an increase in heme oxygenase-1 immunoreactivity and interleukin-10 expression. These results suggest that CAPE exerts a remarkable neuroprotective effect on ischemic brain injury via its anti-inflammatory properties, thereby providing a benefit to the therapy of cerebral infarction.
Collapse
Affiliation(s)
- Sun Ae Hwang
- Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Chi Dae Kim
- Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Won Suk Lee
- Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| |
Collapse
|
131
|
Sun P, Hernandez-Guillamón M, Campos-Martorell M, Simats A, Montaner J, Unzeta M, Solé M. Simvastatin blocks soluble SSAO/VAP-1 release in experimental models of cerebral ischemia: Possible benefits for stroke-induced inflammation control. Biochim Biophys Acta Mol Basis Dis 2017; 1864:542-553. [PMID: 29175057 DOI: 10.1016/j.bbadis.2017.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/19/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
Beyond cholesterol reduction, statins mediate their beneficial effects on stroke patients through pleiotropic actions. They have shown anti-inflammatory properties by a number of different mechanisms, including the inhibition of NF-κB transcriptional activity and the consequent increase and release of adhesion molecules. We have studied simvastatin's effects on the vascular enzyme semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1), which is involved in stroke-mediated brain injury. SSAO/VAP-1 has leukocyte-binding capacity and mediates the expression of other adhesion proteins through signaling molecules generated by its catalytic activity. Our results indicate that soluble SSAO/VAP-1 is released into the bloodstream after an ischemic stimulus, in parallel with an increase in E-selectin and VCAM-1 and correlating with infarct volume. Simvastatin blocks soluble SSAO/VAP-1 release and prevents E-selectin and VCAM-1 overexpression as well. Simvastatin also effectively blocks SSAO/VAP-1-mediated leukocyte adhesion, although it is not an enzymatic inhibitor of SSAO in vitro. In addition, simvastatin-induced changes in adhesion molecules are greater in human brain endothelial cell cultures expressing SSAO/VAP-1, compared to those not expressing it, indicating some synergic effect with SSAO/VAP-1. We think that part of the beneficial effect of simvastatin in stroke is mediated by the attenuation of the SSAO/VAP-1-dependent inflammatory response.
Collapse
Affiliation(s)
- Ping Sun
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mar Hernandez-Guillamón
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Campos-Martorell
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Simats
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mercedes Unzeta
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| | - Montse Solé
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| |
Collapse
|
132
|
Evans NR, Tarkin JM, Buscombe JR, Markus HS, Rudd JHF, Warburton EA. PET imaging of the neurovascular interface in cerebrovascular disease. Nat Rev Neurol 2017; 13:676-688. [PMID: 28984315 DOI: 10.1038/nrneurol.2017.129] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cerebrovascular disease encompasses a range of pathologies that affect different components of the cerebral vasculature and brain parenchyma. Large artery atherosclerosis, acute cerebral ischaemia, and intracerebral small vessel disease all demonstrate altered metabolic processes that are key to their pathogenesis. Although structural imaging techniques such as MRI are the mainstay of clinical care and research in cerebrovascular disease, they have limited ability to detect these pathophysiological processes in vivo. By contrast, PET can detect and quantify metabolic processes that are relevant to each facet of cerebrovascular disease. Information obtained from PET studies has helped to shape the understanding of key concepts in cerebrovascular medicine, including vulnerable atherosclerotic plaque, salvageable ischaemic penumbra, neuroinflammation and selective neuronal loss after ischaemic insult. PET has also helped to elucidate the relationships between chronic hypoxia, neuroinflammation, and amyloid-β deposition in cerebral small vessel disease. This Review describes how PET-based imaging of metabolic processes at the neurovascular interface has contributed to our understanding of cerebrovascular disease.
Collapse
Affiliation(s)
- Nicholas R Evans
- Department of Clinical Neurosciences, University of Cambridge, Box 83, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - John R Buscombe
- Department of Nuclear Medicine, Box 219, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Box 83, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Elizabeth A Warburton
- Department of Clinical Neurosciences, University of Cambridge, Box 83, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
133
|
Ding P, Ren D, He S, He M, Zhang G, Chen Y, Sang H, Peng Z, Yan W. Sirt1 mediates improvement in cognitive defects induced by focal cerebral ischemia following hyperbaric oxygen preconditioning in rats. Physiol Res 2017; 66:1029-1039. [PMID: 28937253 DOI: 10.33549/physiolres.933544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hyperbaric oxygen preconditioning (HBO-PC) has been proposed as a safe and practical approach for neuroprotection in ischemic stroke. However, it is not known whether HPO-PC can improve cognitive deficits induced by cerebral ischemia, and the mechanistic basis for any beneficial effects remains unclear. We addressed this in the present study using rats subjected to middle cerebral artery occlusion (MCAO) as an ischemic stroke model following HBO-PC. Cognitive function and expression of phosphorylated neurofilament heavy polypeptide (pNF-H) and doublecortin (DCX) in the hippocampus were evaluated 14 days after reperfusion and after short interfering RNA-mediated knockdown of sirtuin1 (Sirt1). HBO-PC increased pNF-H and DCX expression and mitigated cognitive deficits in MCAO rats. However, these effects were abolished by Sirt1 knockdown. Our results suggest that HBO-PC can protect the brain from injury caused by ischemia-reperfusion and that Sirt1 is a potential molecular target for therapeutic approaches designed to minimize cognitive deficits caused by cerebral ischemia.
Collapse
Affiliation(s)
- P Ding
- Department of Anesthesiology, Gansu Provincial Hospital, Lanzhou, China, Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China. pengzhengwu1446@ 163.com and
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Wang X, Wang C, Yang Y, Ni J. New monocyte locomotion inhibitory factor analogs protect against cerebral ischemia-reperfusion injury in rats. Bosn J Basic Med Sci 2017; 17:221-227. [PMID: 28273031 DOI: 10.17305/bjbms.2017.1622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/13/2016] [Accepted: 10/16/2016] [Indexed: 12/25/2022] Open
Abstract
Monocyte locomotion inhibitory factor (MLIF) is an oligopeptide with anti-inflammatory properties. The carboxyl-terminal end group Cys-Asn-Ser serves as the pharmacophore of MLIF. The aim of this study was to investigate the neuroprotective effects of two new synthetic analogs, Arg-Cys-Asn-Ser and D-Cys-Asn-Ser, on focal cerebral ischemia, which were designed and synthesized to increase the penetrability and enzymatic stability of Cys-Asn-Ser. Ninety-one male Sprague-Dawley rats were randomly divided into six groups: I - Sham; II - Ischemia-reperfusion (I/R); III - Nimodipine; IV - Cys-Asn-Ser; V - D-Cys-Asn-Ser; and VI - Arg-Cys-Asn-Ser. The rats in groups II-VI were subjected to middle cerebral artery occlusion. After 24 hours of reperfusion, the neurological deficit, cerebral infarct volume, and levels of the pro-inflammatory factors interleukin-1β (IL-1β) and tumor necrosis factor-alpha in brain tissue homogenates were assessed. Compared with the sham group, the mean neurological deficit scores were significantly higher in groups II-VI (p ≤ 0.019 for all). The mean infarct volumes were significantly higher in I/R and Cys-Asn-Ser groups compared with the sham group (both p ≤ 0.046). The mean IL-1β level was significantly lower in D-Cys-Asn-Ser and Arg-Cys-Asn-Ser groups compared with I/R group (both p ≤ 0.046). In conclusion, the results showed that Arg-Cys-Asn-Ser and D-Cys-Asn-Ser have the potential for protective effects against focal cerebral ischemia injury.
Collapse
Affiliation(s)
- Xiaoping Wang
- Department of Pharmacy, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Department of Respiratory Medicine, The Second Hospital of Lanzhou University, Lanzhou, China.
| | | | | | | |
Collapse
|
135
|
Cheon SY, Kim EJ, Kim JM, Kam EH, Ko BW, Koo BN. Regulation of Microglia and Macrophage Polarization via Apoptosis Signal-Regulating Kinase 1 Silencing after Ischemic/Hypoxic Injury. Front Mol Neurosci 2017; 10:261. [PMID: 28855861 PMCID: PMC5557792 DOI: 10.3389/fnmol.2017.00261] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/31/2017] [Indexed: 12/28/2022] Open
Abstract
Inflammation is implicated in ischemic stroke and is involved in abnormal homeostasis. Activation of the immune system leads to breakdown of the blood-brain barrier and, thereby, infiltration of immune cells into the brain. Upon cerebral ischemia, infiltrated macrophages and microglia (resident CNS immune cell) are activated, change their phenotype to M1 or M2 based on the microenvironment, migrate toward damaged tissue, and are involved in repair or damage. Those of M1 phenotype release pro-inflammatory mediators, which are associated with tissue damage, while those of M2 phenotype release anti-inflammatory mediators, which are related to tissue recovery. Moreover, late inflammation continually stimulates immune cell infiltration and leads to brain infarction. Therefore, regulation of M1/M2 phenotypes under persistent inflammatory conditions after cerebral ischemia is important for brain repair. Herein, we focus on apoptosis signal-regulating kinase 1 (ASK1), which is involved in apoptotic cell death, brain infarction, and production of inflammatory mediators after cerebral ischemia. We hypothesized that ASK1 is involved in the polarization of M1/M2 phenotype and the function of microglia and macrophage during the late stage of ischemia/hypoxia. We investigated the effects of ASK1 in mice subjected to middle cerebral artery occlusion and on BV2 microglia and RAW264.7 macrophage cell lines subjected to oxygen-glucose deprivation. Our results showed that ASK1 silencing effectively reduced Iba-1 or CD11b-positive cells in ischemic areas, suppressed pro-inflammatory cytokines, and increased anti-inflammatory mediator levels at 7 days after cerebral ischemia. In cultured microglia and macrophages, ASK1 inhibition, induced by NQDI-1 drug, decreased the expression and release of M1-associated factors and increased those of M2-associated factors after hypoxia/reperfusion (H/R). At the gene level, ASK1 inhibition suppressed M1-associated genes and augmented M2-associated genes. In gap closure assay, ASK1 inhibition reduced the migration rate of microglia and macrophages after H/R. Taken together, our results provide new information that suggests ASK1 controls the polarization of M1/M2 and the function of microglia and macrophage under sustained-inflammatory conditions. Regulation of persistent inflammation via M1/M2 polarization by ASK1 is a novel strategy for repair after ischemic stroke.
Collapse
Affiliation(s)
- So Yeong Cheon
- Department of Anesthesiology and Pain Medicine, Yonsei University College of MedicineSeoul, South Korea.,Anesthesia and Pain Research Institute, Yonsei University College of MedicineSeoul, South Korea
| | - Eun Jung Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of MedicineSeoul, South Korea.,Anesthesia and Pain Research Institute, Yonsei University College of MedicineSeoul, South Korea
| | - Jeong Min Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of MedicineSeoul, South Korea.,Anesthesia and Pain Research Institute, Yonsei University College of MedicineSeoul, South Korea
| | - Eun Hee Kam
- Department of Anesthesiology and Pain Medicine, Yonsei University College of MedicineSeoul, South Korea.,Anesthesia and Pain Research Institute, Yonsei University College of MedicineSeoul, South Korea
| | - Byung Woong Ko
- Department of Anesthesiology and Pain Medicine, Yonsei University College of MedicineSeoul, South Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Yonsei University College of MedicineSeoul, South Korea.,Anesthesia and Pain Research Institute, Yonsei University College of MedicineSeoul, South Korea
| |
Collapse
|
136
|
Campbell RA, Vieira-de-Abreu A, Rowley JW, Franks ZG, Manne BK, Rondina MT, Kraiss LW, Majersik JJ, Zimmerman GA, Weyrich AS. Clots Are Potent Triggers of Inflammatory Cell Gene Expression: Indications for Timely Fibrinolysis. Arterioscler Thromb Vasc Biol 2017; 37:1819-1827. [PMID: 28775073 DOI: 10.1161/atvbaha.117.309794] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 07/21/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Blood vessel wall damage often results in the formation of a fibrin clot that traps inflammatory cells, including monocytes. The effect of clot formation and subsequent lysis on the expression of monocyte-derived genes involved in the development and progression of ischemic stroke and other vascular diseases, however, is unknown. Determine whether clot formation and lysis regulates the expression of human monocyte-derived genes that modulate vascular diseases. APPROACH AND RESULTS We performed next-generation RNA sequencing on monocytes extracted from whole blood clots and using a purified plasma clot system. Numerous mRNAs were differentially expressed by monocytes embedded in clots compared with unclotted controls, and IL-8 (interleukin 8) and MCP-1 (monocyte chemoattractant protein-1) were among the upregulated transcripts in both models. Clotted plasma also increased expression of IL-8 and MCP-1, which far exceeded responses observed in lipopolysaccharide-stimulated monocytes. Upregulation of IL-8 and MCP-1 occurred in a thrombin-independent but fibrin-dependent manner. Fibrinolysis initiated shortly after plasma clot formation (ie, 1-2 hours) reduced the synthesis of IL-8 and MCP-1, whereas delayed fibrinolysis was far less effective. Consistent with these in vitro models, monocytes embedded in unresolved thrombi from patients undergoing thrombectomy stained positively for IL-8 and MCP-1. CONCLUSIONS These findings demonstrate that clots are potent inducers of monocyte gene expression and that timely fibrinolysis attenuates inflammatory responses, specifically IL-8 and MCP-1. Dampening of inflammatory gene expression by timely clot lysis may contribute to the clinically proven efficacy of fibrinolytic drug treatment within hours of stroke onset.
Collapse
Affiliation(s)
- Robert A Campbell
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City.
| | - Adriana Vieira-de-Abreu
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Jesse W Rowley
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Zechariah G Franks
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Bhanu Kanth Manne
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Matthew T Rondina
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Larry W Kraiss
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Jennifer J Majersik
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Guy A Zimmerman
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| | - Andrew S Weyrich
- From the Program in Molecular Medicine (R.A.C., J.W.R., Z.G.F., B.K.M., M.T.R., L.W.K., A.S.W.) and Departments of Internal Medicine (R.A.C., A.V.-d.-A., J.W.R., M.T.R., G.A.Z., A.S.W.), Surgery (L.W.K.), and Neurology (J.J.M.), University of Utah, Salt Lake City
| |
Collapse
|
137
|
Tikamdas R, Singhal S, Zhang P, Smith JA, Krause EG, Stevens SM, Song S, Liu B. Ischemia-responsive protein 94 is a key mediator of ischemic neuronal injury-induced microglial activation. J Neurochem 2017. [PMID: 28640931 DOI: 10.1111/jnc.14111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuroinflammation, especially activation of microglia, the key immune cells in the brain, has been proposed to contribute to the pathogenesis of ischemic stroke. However, the dynamics and the potential mediators of microglial activation following ischemic neuronal injury are not well understood. In this study, using oxygen/glucose deprivation and reoxygenation with neuronal and microglial cell cultures as an in vitro model of ischemic neuronal injury, we set out to identify neuronal factors released from injured neurons that are capable of inducing microglial activation. Conditioned media (CM) from hippocampal and cortical neurons exposed to oxygen/glucose deprivation and reoxygenation induced significant activation of microglial cells as well as primary microglia, evidenced by up-regulation of inducible nitric oxide synthase, increased production of nitrite and reactive oxygen species, and increased expression of microglial markers. Mechanistically, neuronal ischemia-responsive protein 94 (Irp94) was a key contributor to microglial activation since significant increase in Irp94 was detected in the neuronal CM following ischemic insult and immunodepletion of Irp94 rendered ischemic neuronal CM ineffective in inducing microglial activation. Ischemic insult-augmented oxidative stress was a major facilitator of neuronal Irp94 release, and pharmacological inhibition of NADPH oxidase significantly reduced the ischemic injury-induced neuronal reactive oxygen species production and Irp94 release. Taken together, these results indicate that neuronal Irp94 may play a pivotal role in the propagation of ischemic neuronal damage. Continued studies may help identify Irp94 and/or related proteins as potential therapeutic targets and/or diagnostic/prognostic biomarkers for managing ischemia-associated brain disorders.
Collapse
Affiliation(s)
- Rajiv Tikamdas
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Sarthak Singhal
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Ping Zhang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Justin A Smith
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Bin Liu
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
138
|
Biliverdin administration ameliorates cerebral ischemia reperfusion injury in rats and is associated with proinflammatory factor downregulation. Exp Ther Med 2017; 14:671-679. [PMID: 28672984 PMCID: PMC5488602 DOI: 10.3892/etm.2017.4549] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 03/17/2017] [Indexed: 02/05/2023] Open
Abstract
Biliverdin (BV), one of the heme oxygenase-1 (HO-1) catalytic products, has been demonstrated to have protective effects in liver ischemia reperfusion injury (IRI). The present study aimed to explore the effects of BV on cerebral IRI, and to investigate the potential mechanisms thereof. Adult male SD rats, weighing 200-240 g, were randomly divided into sham (group S), cerebral ischemia reperfusion control (group C) and BV (group BV) groups. Rats in group C underwent transient middle cerebral artery occlusion (tMCAO) and received 2 ml normal saline; rats in group BV received BV (35 mg/kg) intraperitoneally 15 min prior to reperfusion and 4 h after reperfusion, then twice a day thereafter for 5 days. Group S served as the control. Neurological Severity Scores (NSS) were evaluated at days 1-5 following reperfusion. Staining with 2, 3, 5-triphenyltetrazolium chloride was performed to determine the cerebral infarction at 48 h post reperfusion. mRNA expression levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, inducible nitric oxide synthase (iNOS) and HO-1 in the ischemic cerebral cortex were detected via reverse transcription-quantitative polymerase chain reaction at 3, 6, 12 and 24 h after reperfusion. Western blotting was used to detect the protein expression levels at 3 h after reperfusion. Compared with group S, the NSS, cerebral infarct volume, and the mRNA and protein expression levels of TNF-α, IL-6, IL-1β, iNOS and HO-1 of Group C were significantly increased (P<0.05). However, BV administration significantly improved and reduced these expression levels (P<0.01). The present study indicates that BV is able to ameliorate cerebral IRI in rats and that the mechanism may be associated with the downregulation of proinflammatory factors.
Collapse
|
139
|
Gao XJ, Xie GN, Liu L, Fu ZJ, Zhang ZW, Teng LZ. Sesamol attenuates oxidative stress, apoptosis and inflammation in focal cerebral ischemia/reperfusion injury. Exp Ther Med 2017; 14:841-847. [PMID: 28673008 DOI: 10.3892/etm.2017.4550] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/21/2016] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to evaluate the therapeutic potential of sesamol treatment on focal ischemia/reperfusion (I/R) injury in the rat brain. The results demonstrated that pretreatment with sesamol seven days prior to focal cerebral I/R injury had significant positive effects, including improvements in neurological deficits (P<0.05), and a reduction in malondialdehyde content and elevation of antioxidant levels (superoxide dismutase, glutathione and glutatione peroxidase; both P<0.05). Furthermore, levels of B cell lymphoma-2 (Bcl-2)-associated X protein and caspase-3 were significantly downregulated, whereas the level of Bcl-2 was effectively increased. Conversely, the mRNA expression of proinflammatory cytokines were significantly reduced in focal cerebral I/R injury rats upon sesamol intervention. Therefore, the beneficial effects of sesamol on cerebral I/R injury may be due to the reduction of oxidative stress, inhibition of apoptosis and inflammation. The findings of the present study suggest that sesamol supplementation may serve as potent adjuvant in the treatment of focal cerebral ischemia/reperfusion injury due to its neuroprotective effects.
Collapse
Affiliation(s)
- Xiu-Juan Gao
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China.,Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Guan-Nan Xie
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Lei Liu
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China.,Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Zhi-Jian Fu
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zong-Wang Zhang
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Liang-Zhu Teng
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
140
|
Khoshnam SE, Winlow W, Farbood Y, Moghaddam HF, Farzaneh M. Emerging Roles of microRNAs in Ischemic Stroke: As Possible Therapeutic Agents. J Stroke 2017; 19:166-187. [PMID: 28480877 PMCID: PMC5466283 DOI: 10.5853/jos.2016.01368] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 02/08/2017] [Accepted: 02/27/2017] [Indexed: 01/06/2023] Open
Abstract
Stroke is one of the leading causes of death and physical disability worldwide. The consequences of stroke injuries are profound and persistent, causing in considerable burden to both the individual patient and society. Current treatments for ischemic stroke injuries have proved inadequate, partly owing to an incomplete understanding of the cellular and molecular changes that occur following ischemic stroke. MicroRNAs (miRNA) are endogenously expressed RNA molecules that function to inhibit mRNA translation and have key roles in the pathophysiological processes contributing to ischemic stroke injuries. Potential therapeutic areas to compensate these pathogenic processes include promoting angiogenesis, neurogenesis and neuroprotection. Several miRNAs, and their target genes, are recognized to be involved in these recoveries and repair mechanisms. The capacity of miRNAs to simultaneously regulate several target genes underlies their unique importance in ischemic stroke therapeutics. In this Review, we focus on the role of miRNAs as potential diagnostic and prognostic biomarkers, as well as promising therapeutic agents in cerebral ischemic stroke.
Collapse
Affiliation(s)
- Seyed Esmaeil Khoshnam
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - William Winlow
- Dipartimento di Biologia, Università degli Studi di Napoli, Napoli, Italia.,Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool, UK
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Fathi Moghaddam
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
141
|
Abstract
Stroke is the second most common cause of death and the leading cause of disability worldwide. Brain injury following stroke results from a complex series of pathophysiological events including excitotoxicity, oxidative and nitrative stress, inflammation, and apoptosis. Moreover, there is a mechanistic link between brain ischemia, innate and adaptive immune cells, intracranial atherosclerosis, and also the gut microbiota in modifying the cerebral responses to ischemic insult. There are very few treatments for stroke injuries, partly owing to an incomplete understanding of the diverse cellular and molecular changes that occur following ischemic stroke and that are responsible for neuronal death. Experimental discoveries have begun to define the cellular and molecular mechanisms involved in stroke injury, leading to the development of numerous agents that target various injury pathways. In the present article, we review the underlying pathophysiology of ischemic stroke and reveal the intertwined pathways that are promising therapeutic targets.
Collapse
|
142
|
Wu L, Wang HM, Li JL, Feng HX, Zhao WM, Zhang HY. Dual anti-ischemic effects of rosmarinic acid n-butyl ester via alleviation of DAPK-p53-mediated neuronal damage and microglial inflammation. Acta Pharmacol Sin 2017; 38:459-468. [PMID: 28216621 PMCID: PMC5533207 DOI: 10.1038/aps.2016.156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/10/2016] [Indexed: 12/18/2022] Open
Abstract
The discovery of efficacious anti-ischemic drugs remains a challenge. Recently we have found that rosmarinic acid n-butyl ester (RABE), a derivative of rosmarinic acid, significantly protects SH-SY5Y cells against oxygen glucose deprivation (OGD)-induced cell death. In the present study we simultaneously investigated the effects of RABE on the two key players in the pathophysiology of cerebral ischemia, ischemic neuronal damage and microglial inflammation. Pretreatment with RABE (1, 10 μmol/L) dose-dependently attenuated OGD- or H2O2-induced reduction of the viability of SH-SY5Y neuroblastoma cells. RABE pretreatment concurrently reduced the apoptotic cell rate, down-regulated the expression of the pro-apoptotic proteins Bax and p53, and up-regulated the expression of the anti-apoptotic protein phosphorylated death-associated protein kinase (DAPK). Furthermore, pretreatment with RABE (3 μmol/L) markedly inhibited lipopolysaccharide (LPS)-induced increases in the release of TNF-α, IL-1β, NO and PGE2, and the expression levels of iNOS, and COX-2 in cultured rat microglial cells. In conclusion, these results reveal for the first time the potential anti-ischemic effects of RABE on neuronal and glial cells and elucidate the molecular mechanisms involved in its dual beneficial profiles in vitro. RABE may be a promising drug lead/candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lei Wu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong-min Wang
- Department of Natural Product Chemistry and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin-long Li
- Department of Natural Product Chemistry and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong-xuan Feng
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei-min Zhao
- Department of Natural Product Chemistry and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hai-yan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
143
|
Zeng WX, Han YL, Zhu GF, Huang LQ, Deng YY, Wang QS, Jiang WQ, Wen MY, Han QP, Xie D, Zeng HK. Hypertonic saline attenuates expression of Notch signaling and proinflammatory mediators in activated microglia in experimentally induced cerebral ischemia and hypoxic BV-2 microglia. BMC Neurosci 2017; 18:32. [PMID: 28288585 PMCID: PMC5348816 DOI: 10.1186/s12868-017-0351-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/08/2017] [Indexed: 11/30/2022] Open
Abstract
Background Ischemic stroke is a major disease that threatens human health in ageing population. Increasing evidence has shown that neuroinflammatory mediators play crucial roles in the pathophysiology of cerebral ischemia injury. Notch signaling is recognized as the cell fate signaling but recent evidence indicates that it may be involved in the inflammatory response in activated microglia in cerebral ischemia. Previous report in our group demonstrated hypertonic saline (HS) could reduce the release of interleukin-1 beta and tumor necrosis factor-alpha in activated microglia, but the underlying molecular and cellular mechanisms have remained uncertain. This study was aimed to explore whether HS would partake in regulating production of proinflammatory mediators through Notch signaling. Results HS markedly attenuated the expression of Notch-1, NICD, RBP-JK and Hes-1 in activated microglia both in vivo and in vitro. Remarkably, HS also reduced the expression of iNOS in vivo, while the in vitro levels of inflammatory mediators Phos-NF-κB, iNOS and ROS were reduced by HS as well. Conclusion Our results suggest that HS may suppress of inflammatory mediators following ischemia/hypoxic through the Notch signaling which operates synergistically with NF-κB pathway in activated microglia. Our study has provided the morphological and biochemical evidence that HS can attenuate inflammation reaction and can be neuroprotective in cerebral ischemia, thus supporting the use of hypertonic saline by clinicians in patients with an ischemia stroke. Electronic supplementary material The online version of this article (doi:10.1186/s12868-017-0351-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen-Xin Zeng
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Yong-Li Han
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.
| | - Gao-Feng Zhu
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Lin-Qiang Huang
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Yi-Yu Deng
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Qiao-Sheng Wang
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Wen-Qiang Jiang
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Miao-Yun Wen
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Qian-Peng Han
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Di Xie
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Hong-Ke Zeng
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
144
|
Tu TM, Kolls BJ, Soderblom EJ, Cantillana V, Ferrell PD, Moseley MA, Wang H, Dawson HN, Laskowitz DT. Apolipoprotein E mimetic peptide, CN-105, improves outcomes in ischemic stroke. Ann Clin Transl Neurol 2017; 4:246-265. [PMID: 28382306 PMCID: PMC5376751 DOI: 10.1002/acn3.399] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/09/2017] [Accepted: 02/06/2017] [Indexed: 01/05/2023] Open
Abstract
Objective At present, the absence of a pharmacological neuroprotectant represents an important unmet clinical need in the treatment of ischemic and traumatic brain injury. Recent evidence suggests that administration of apolipoprotein E mimetic therapies represent a viable therapeutic strategy in this setting. We investigate the neuroprotective and anti‐inflammatory properties of the apolipoprotein E mimetic pentapeptide, CN‐105, in a microglial cell line and murine model of ischemic stroke. Methods Ten to 13‐week‐old male C57/BL6 mice underwent transient middle cerebral artery occlusion and were randomly selected to receive CN‐105 (0.1 mg/kg) in 100 μL volume or vehicle via tail vein injection at various time points. Survival, motor‐sensory functional outcomes using rotarod test and 4‐limb wire hanging test, infarct volume assessment using 2,3,5‐Triphenyltetrazolium chloride staining method, and microglial activation in the contralateral hippocampus using F4/80 immunostaining were assessed at various time points. In vitro assessment of tumor necrosis factor‐alpha secretion in a microglial cell line was performed, and phosphoproteomic analysis conducted to explore early mechanistic pathways of CN‐105 in ischemic stroke. Results Mice receiving CN‐105 demonstrated improved survival, improved functional outcomes, reduced infarct volume, and reduced microglial activation. CN‐105 also suppressed inflammatory cytokines secretion in microglial cells in vitro. Phosphoproteomic signals suggest that CN‐105 reduces proinflammatory pathways and lower oxidative stress. Interpretation CN‐105 improves functional and histological outcomes in a murine model of ischemic stroke via modulation of neuroinflammatory pathways. Recent clinical trial of this compound has demonstrated favorable pharmacokinetic and safety profile, suggesting that CN‐105 represents an attractive candidate for clinical translation.
Collapse
Affiliation(s)
- Tian Ming Tu
- Department of Neurology Duke University School of Medicine Durham North Carolina; Department of Neurology National Neuroscience Institute Tan Tock Seng Campus Singapore
| | - Brad J Kolls
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Erik J Soderblom
- Duke Proteomics Core Facility Center for Genomic and Computational Biology Duke University Durham North Carolina
| | - Viviana Cantillana
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Paul Durham Ferrell
- Department of Pathology Duke University School of Medicine Durham North Carolina
| | - M Arthur Moseley
- Duke Proteomics Core Facility Center for Genomic and Computational Biology Duke University Durham North Carolina
| | - Haichen Wang
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Hana N Dawson
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Daniel T Laskowitz
- Department of Neurology Duke University School of Medicine Durham North Carolina
| |
Collapse
|
145
|
Semax, an analog of ACTH (4-7), regulates expression of immune response genes during ischemic brain injury in rats. Mol Genet Genomics 2017; 292:635-653. [PMID: 28255762 DOI: 10.1007/s00438-017-1297-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 02/07/2017] [Indexed: 12/20/2022]
Abstract
Brain stroke continues to claim the lives of million people every year. To build the effective strategies for stroke treatment it is necessary to understand the neuroprotective mechanisms that are able to prevent the ischemic injury. Consisting of the ACTH(4-7) fragment and the tripeptide Pro-Gly-Pro (PGP), the synthetic peptide Semax effectively protects brain against ischemic stroke. However, the molecular mechanisms underlying its neuroprotection and participation of PGP in them are still needed to be clarified. To reveal biological processes and signaling pathways, which are affected by Semax and PGP, we performed the transcriptome analysis of cerebral cortex of rats with focal cerebral ischemia treated by these peptides. The genome-wide biochip data analysis detected the differentially expressed genes (DEGs) and bioinformatic web-tool Ingenuity iReport found DEGs associations with several biological processes and signaling pathways. The immune response is the process most markedly affected by the peptide: Semax enhances antigen presentation signaling pathway, intensifies the effect of ischemia on the interferon signaling pathways and affects the processes for synthesizing immunoglobulins. Semax significantly increased expression of the gene encoding the immunoglobulin heavy chain, highly affects on cytokine, stress response and ribosomal protein-encoding genes after occlusion. PGP treatment of rats with ischemia attenuates the immune activity and suppresses neurotransmission in the CNS. We suppose that neuroprotective mechanism of Semax is realized via the neuroimmune crosstalk, and the new properties of PGP were found under ischemia. Our results provided the basis for further proteomic investigations in the field of searching Semax neuroprotection mechanism.
Collapse
|
146
|
Lee JH, Kam EH, Kim JM, Kim SY, Kim EJ, Cheon SY, Koo BN. Intranasal Administration of Interleukin-1 Receptor Antagonist in a Transient Focal Cerebral Ischemia Rat Model. Biomol Ther (Seoul) 2017; 25:149-157. [PMID: 27530114 PMCID: PMC5340539 DOI: 10.4062/biomolther.2016.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/06/2016] [Accepted: 05/24/2016] [Indexed: 11/05/2022] Open
Abstract
The interleukin-1 receptor antagonist (IL-1RA) is a potential stroke treatment candidate. Intranasal delivery is a novel method thereby a therapeutic protein can be penetrated into the brain parenchyma by bypassing the blood-brain barrier. Thus, this study tested whether intranasal IL-1RA can provide neuroprotection and brain penetration in transient cerebral ischemia. In male Sprague-Dawley rats, focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) for 1 h. The rats simultaneously received 50 mg/kg human IL-1RA through the intranasal (IN group) or intraperitoneal route (IP group). The other rats were given 0.5 mL/kg normal saline (EC group). Neurobehavioral function, infarct size, and the concentration of the administered human IL-1RA in the brain tissue were assessed. In addition, the cellular distribution of intranasal IL-1RA in the brain and its effect on proinflammatory cytokines expression were evaluated. Intranasal IL-1RA improved neurological deficit and reduced infarct size until 7 days after MCAO (p<0.05). The concentrations of the human IL-1RA in the brain tissue 24 h after MCAO were significantly greater in the IN group than in the IP group (p<0.05). The human IL-1RA was confirmed to be co-localized with neuron and microglia. Furthermore, the IN group had lower expression of interleukin-1β and tumor necrosis factor-α at 6 h after MCAO than the EC group (p<0.05). These results suggest that intranasal IL-1RA can reach the brain parenchyma more efficiently and provide superior neuroprotection in the transient focal cerebral ischemia.
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eun Hee Kam
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeong Min Kim
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - So Yeon Kim
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eun Jeong Kim
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - So Yeong Cheon
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
147
|
Qu Y, Zhang HL, Zhang XP, Jiang HL. Arachidonic acid attenuates brain damage in a rat model of ischemia/reperfusion by inhibiting inflammatory response and oxidative stress. Hum Exp Toxicol 2017; 37:135-141. [PMID: 29233001 DOI: 10.1177/0960327117692134] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to study the effects of arachidonic acid (ARA) in a rat brain ischemia/reperfusion model induced by middle cerebral artery occlusion (MCAO). A total of 50 rats were randomly divided into five groups: control group, MCAO group, MCAO + ARA 0.3 g/kg group, MCAO + ARA 1 g/kg group, and MCAO + ARA 3 g/kg group. The MCAO + ARA groups received ARA by intraperitoneal injection daily for 14 consecutive days, while the rats in the control and MCAO groups were given equivalent volume of saline. We detected the Morris water maze test and pathological changes to investigate the ischemia/reperfusion injury. The protein levels of tumor necrosis factor-alpha and interleukin-6 in the hippocampus were detected by enzyme-linked immunosorbent assay kits. In addition, the activities of superoxide dismutase, glutathione peroxidase, and malondialdehyde were assayed in hippocampus homogenates to evaluate the oxidative stress after ischemia/reperfusion. The results indicated that ARA administration decreased biochemical parameters of inflammation and oxidative stress. Morris water maze test and histopathological examination further verified the protective effects of ARA on ischemia/reperfusion injury rats. These findings demonstrated that ARA could protect MCAO-induced brain injury rats by inhibition of inflammation and oxidative stress, suggesting that it may have potential as a therapy for cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Y Qu
- 1 Department of Emergency, Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - H-L Zhang
- 2 Department of Neurology, Yuhuangding Hospital, Qingdao University, Yantai, China
| | - X-P Zhang
- 3 Department of Pharmacy, Yuhuangding Hospital, Qingdao University, Yantai, China
| | - H-L Jiang
- 4 Kidney Disease/Blood Purification, Jinan Central Hospital, Jinan, China
| |
Collapse
|
148
|
Jin Q, Cai Y, Li S, Liu H, Zhou X, Lu C, Gao X, Qian J, Zhang J, Ju S, Li C. Edaravone-Encapsulated Agonistic Micelles Rescue Ischemic Brain Tissue by Tuning Blood-Brain Barrier Permeability. Am J Cancer Res 2017; 7:884-898. [PMID: 28382161 PMCID: PMC5381251 DOI: 10.7150/thno.18219] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 11/23/2016] [Indexed: 12/31/2022] Open
Abstract
Thrombolysis has been a standard treatment for ischemic stroke. However, only 2-7% patients benefit from it because the thrombolytic agent has to be injected within 4.5 h after the onset of symptoms to avoid the increasing risk of intracerebral hemorrhage. As the only clinically approved neuroprotective drug, edaravone (EDV) rescues ischemic brain tissues by eradicating over-produced reactive oxygen species (ROS) without the limitation of therapeutic time-window. However, EDV's short circulation half-life and inadequate cerebral uptake attenuate its therapeutic efficacy. Here we developed an EDV-encapsulated agonistic micelle (EDV-AM) to specifically deliver EDV into brain ischemia by actively tuning blood-brain barrier (BBB) permeability. The EDV-AM actively up-regulated endothelial monolayer permeability in vitro. HPLC studies showed that EDV-AM delivered more EDV into brain ischemia than free EDV after intravenous injection. Magnetic resonance imaging also demonstrated that EDV-AM more rapidly salvaged ischemic tissue than free EDV. Diffusion tensor imaging indicated the highest efficiency of EDV-AM in accelerating axonal remodeling in the ipsilesional white matter and improving functional behaviors of ischemic stroke models. The agonistic micelle holds promise to improve the therapeutic efficiency of ischemic stroke patients who miss the thrombolytic treatment.
Collapse
|
149
|
Shukla V, Shakya AK, Perez-Pinzon MA, Dave KR. Cerebral ischemic damage in diabetes: an inflammatory perspective. J Neuroinflammation 2017; 14:21. [PMID: 28115020 PMCID: PMC5260103 DOI: 10.1186/s12974-016-0774-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/07/2016] [Indexed: 12/16/2022] Open
Abstract
Stroke is one of the leading causes of death worldwide. A strong inflammatory response characterized by activation and release of cytokines, chemokines, adhesion molecules, and proteolytic enzymes contributes to brain damage following stroke. Stroke outcomes are worse among diabetics, resulting in increased mortality and disabilities. Diabetes involves chronic inflammation manifested by reactive oxygen species generation, expression of proinflammatory cytokines, and activation/expression of other inflammatory mediators. It appears that increased proinflammatory processes due to diabetes are further accelerated after cerebral ischemia, leading to increased ischemic damage. Hypoglycemia is an intrinsic side effect owing to glucose-lowering therapy in diabetics, and is known to induce proinflammatory changes as well as exacerbate cerebral damage in experimental stroke. Here, we present a review of available literature on the contribution of neuroinflammation to increased cerebral ischemic damage in diabetics. We also describe the role of hypoglycemia in neuroinflammation and cerebral ischemic damage in diabetics. Understanding the role of neuroinflammatory mechanisms in worsening stroke outcome in diabetics may help limit ischemic brain injury and improve clinical outcomes.
Collapse
Affiliation(s)
- Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA.,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA
| | - Akhalesh Kumar Shakya
- Present address: Department of Microbiology and Immunology, and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA.,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA. .,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
150
|
Chen Y, Li Y, Xu H, Li G, Ma Y, Pang YJ. MORIN MITIGATES OXIDATIVE STRESS, APOPTOSIS AND INFLAMMATION IN CEREBRAL ISCHEMIC RATS. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2017; 14:348-355. [PMID: 28573251 PMCID: PMC5446461 DOI: 10.21010/ajtcam.v14i2.36] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: Morin is a flavanoid which exhibits potent antioxidant activity in various oxidative stress related diseases. The current study was attempted to scrutinize the preclinical bio-efficacy of morin on focal ischemia. Methods: The animal model of focal cerebral ischemic injury was done by midbrain carotid artery occlusion (MCAO) method, followed by Morin (30mg/kg) administration for seven days. Results: The outcome of the study showed that treatment with morin displayed positive effects in reducing the focal cerebral ischemia. This effect was evident with the improvements in neurological deficits, reduction in MDA content and elevation of antioxidant levels (SOD, GSH and Gpx). Furthermore, protein expression of Bax and caspase-3 were effectively down-regulated, whilst the expression of Bcl-2 was significantly elevated. On the other hand, the mRNA expression of proinflammatory cytokines was significantly reduced in focal cerebral ischemic rats upon morin intervention. Conclusion: Thus, the beneficial effects of morin on cerebral ischemia assault may result from the reduction of oxidative stress, inhibition of apoptosis and inflammation. The neuroprotective effects of morin supplement may serve as potent adjuvant in the amelioration of ischemic stroke.
Collapse
Affiliation(s)
- Yanrong Chen
- Department of Neurology, Binzhou city central hospital, Binzhou, Shandong 251700, China
| | - Yanke Li
- Department of Neurology, Binzhou city central hospital, Binzhou, Shandong 251700, China
| | - Huali Xu
- Department of Neurology, Binzhou city central hospital, Binzhou, Shandong 251700, China
| | - Gang Li
- Department of Neurology, Binzhou city central hospital, Binzhou, Shandong 251700, China
| | - Yunxia Ma
- Department of Neurology, Binzhou city central hospital, Binzhou, Shandong 251700, China
| | - Yu Jun Pang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|