51
|
Zhang ZQ, Song JY, Jia YQ, Zhang YK. Buyanghuanwu decoction promotes angiogenesis after cerebral ischemia/reperfusion injury: mechanisms of brain tissue repair. Neural Regen Res 2016; 11:435-40. [PMID: 27127482 PMCID: PMC4829008 DOI: 10.4103/1673-5374.179055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Buyanghuanwu decoction has been shown to protect against cerebral ischemia/reperfusion injury, but the underlying mechanisms remain unclear. In this study, rats were intragastrically given Buyanghuanwu decoction, 15 mL/kg, for 3 days. A rat model of cerebral ischemia/reperfusion injury was established by middle cerebral artery occlusion. In rats administered Buyanghuanwu decoction, infarct volume was reduced, serum vascular endothelial growth factor and integrin αvβ3 levels were increased, and brain tissue vascular endothelial growth factor and CD34 expression levels were increased compared with untreated animals. These effects of Buyanghuanwu decoction were partially suppressed by an angiogenesis inhibitor (administered through the lateral ventricle for 7 consecutive days). These data suggest that Buyanghuanwu decoction promotes angiogenesis, improves cerebral circulation, and enhances brain tissue repair after cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Zhen-Qiang Zhang
- Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Jun-Ying Song
- Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Ya-Quan Jia
- Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Yun-Ke Zhang
- First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| |
Collapse
|
52
|
Neuroprotection against cerebral ischemia/reperfusion injury by intravenous administration of liposomal fasudil. Int J Pharm 2016; 506:129-37. [PMID: 27107903 DOI: 10.1016/j.ijpharm.2016.04.046] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/25/2016] [Accepted: 04/15/2016] [Indexed: 12/31/2022]
Abstract
Fasudil, a Rho-kinase inhibitor, is a promising neuroprotectant against ischemic stroke; however, its low bioavailability is an obstacle to be overcome. Our previous study revealed that the liposomal drug delivery system is a hopeful strategy to increase the therapeutic efficacy of neuroprotectants. In the present study, the usefulness of intravenously administered liposomal fasudil for cerebral ischemia/reperfusion (I/R) injury treatment was examined in transient middle cerebral artery occlusion (t-MCAO) rats. The results showed that PEGylated liposomes of approximately 100nm in diameter accumulated more extensively in the I/R region compared with those of over 200nm. Confocal images showed that fluorescence-labeled liposomal fasudil was widely distributed in the I/R region, and was not noticeably taken up by microglia, which are well-known resident macrophages in the brain, and neuronal cells. These data indicated that liposomal fasudil mainly exerted its pharmacological activity by releasing fasudil from the liposomes in the I/R region. Moreover, liposomal fasudil effectively suppressed neutrophil invasion and brain cell damage in the t-MCAO rats, resulting in amelioration of their motor function deficits. These findings demonstrated both the importance of particle size for neuroprotectant delivery and the effectiveness of liposomal fasudil for the treatment of cerebral I/R injury.
Collapse
|
53
|
Yang SI, Yuan Y, Jiao S, Luo QI, Yu J. Calcitonin gene-related peptide protects rats from cerebral ischemia/reperfusion injury via a mechanism of action in the MAPK pathway. Biomed Rep 2016; 4:699-703. [PMID: 27284409 PMCID: PMC4887836 DOI: 10.3892/br.2016.658] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/14/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the protective function and underlying mechanism of calcitonin gene-related peptide (CGRP) on cerebral ischemia/reperfusion damage in rats. Adult male Wistar rats were selected for the establishment of an ischemia/reperfusion injury model through the application of a middle cerebral artery occlusion. Animals were randomly divided into 6 groups of 24 animals. Drugs were administered according to the design of each group; animals were administered CGRP, CGRP8-37, PD98059 and SB20358. The neurobehavioral scores of the rat cerebral ischemia model in each group were calculated. The infarction range of the rat brain tissues was observed by 2,3,5-triphenyltetrazolium chloride staining. The expression levels of three proteins, phosphorylated c-Jun N-terminal kinase (JNK)/JNK, phosphorylated extracellular signal-regulated protein kinase (ERK)/ERK and p-p38/p38, in the mitogen-activated protein kinase (MAPK) pathway in the brain tissues was detected by western blotting. The results showed that CGRP could improve the neurobehavioral function of the ischemic rats and reduce the infarction range. Western blotting results confirmed that the function of the CGRP was mediated mainly through the reduction of the JNK and p38 phosphorylation and the promotion of ERK phosphorylation. Therefore, the present study confirmed that an increase in the exogenous CRGP could effectively improve ischemia/reperfusion injury of the brain tissue. The mechanisms of action were achieved through a reduction in JNK and p38 phosphorylation and an increase in ERL phosphorylation in the MAPK pathway. These mechanisms were interdependent.
Collapse
Affiliation(s)
- S I Yang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China; Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yongjie Yuan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shan Jiao
- Department of Endodontics, Stomatological Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Q I Luo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jinlu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
54
|
Gao S, Mo J, Chen L, Wang Y, Mao X, Shi Y, Zhang X, Yu R, Zhou X. Astrocyte GGTI-mediated Rac1 prenylation upregulates NF-κB expression and promotes neuronal apoptosis following hypoxia/ischemia. Neuropharmacology 2016; 103:44-56. [DOI: 10.1016/j.neuropharm.2015.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/20/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022]
|
55
|
Exogenous S1P Exposure Potentiates Ischemic Stroke Damage That Is Reduced Possibly by Inhibiting S1P Receptor Signaling. Mediators Inflamm 2015; 2015:492659. [PMID: 26576074 PMCID: PMC4630407 DOI: 10.1155/2015/492659] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/18/2015] [Accepted: 09/28/2015] [Indexed: 01/31/2023] Open
Abstract
Initial and recurrent stroke produces central nervous system (CNS) damage, involving neuroinflammation. Receptor-mediated S1P signaling can influence neuroinflammation and has been implicated in cerebral ischemia through effects on the immune system. However, S1P-mediated events also occur within the brain itself where its roles during stroke have been less well studied. Here we investigated the involvement of S1P signaling in initial and recurrent stroke by using a transient middle cerebral artery occlusion/reperfusion (M/R) model combined with analyses of S1P signaling. Gene expression for S1P receptors and involved enzymes was altered during M/R, supporting changes in S1P signaling. Direct S1P microinjection into the normal CNS induced neuroglial activation, implicating S1P-initiated neuroinflammatory responses that resembled CNS changes seen during initial M/R challenge. Moreover, S1P microinjection combined with M/R potentiated brain damage, approximating a model for recurrent stroke dependent on S1P and suggesting that reduction in S1P signaling could ameliorate stroke damage. Delivery of FTY720 that removes S1P signaling with chronic exposure reduced damage in both initial and S1P-potentiated M/R-challenged brain, while reducing stroke markers like TNF-α. These results implicate direct S1P CNS signaling in the etiology of initial and recurrent stroke that can be therapeutically accessed by S1P modulators acting within the brain.
Collapse
|
56
|
Fukuta T, Ishii T, Asai T, Sato A, Kikuchi T, Shimizu K, Minamino T, Oku N. Treatment of stroke with liposomal neuroprotective agents under cerebral ischemia conditions. Eur J Pharm Biopharm 2015; 97:1-7. [PMID: 26455340 DOI: 10.1016/j.ejpb.2015.09.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 08/21/2015] [Accepted: 09/30/2015] [Indexed: 01/09/2023]
Abstract
Since the proportion of patients given thrombolytic therapy with tissue plasminogen activator (t-PA) is very limited because of the narrow therapeutic window, the development of new therapies for ischemic stroke has been desired. We previously reported that liposomes injected intravenously accumulate in the ischemic region of the brain via disruption of the blood-brain barrier that occurs under cerebral ischemia. In the present study, we investigated the efficacy of a liposomal neuroprotective agent in middle cerebral artery occlusion (MCAO) rats to develop ischemic stroke therapy prior to the recovery of cerebral blood flow. For this purpose, PEGylated liposomes encapsulating FK506 (FK506-liposomes) were prepared and injected intravenously into MCAO rats after a 1-h occlusion. This treatment significantly suppressed the expansion of oxidative stress and brain cell damage. In addition, administration of FK506-liposomes before reperfusion significantly ameliorated motor function deficits of the rats caused by ischemia/reperfusion injury. These findings suggest that FK506-liposomes effectively exerted a neuroprotective effect during ischemic conditions, and that combination therapy with a liposomal neuroprotectant plus t-PA could be a promising therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Japan Society for the Promotion of Science (JSPS), 8 Ichiban-cho, Chiyoda-ku, Tokyo 102-8472, Japan
| | - Takayuki Ishii
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Akihiko Sato
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takashi Kikuchi
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kosuke Shimizu
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Tetsuo Minamino
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Naoto Oku
- Department of Medical Biochemistry, University of Shizuoka Graduate School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
57
|
Garbuzova-Davis S, Haller E, Williams SN, Haim ED, Tajiri N, Hernandez-Ontiveros DG, Frisina-Deyo A, Boffeli SM, Sanberg PR, Borlongan CV. Compromised blood-brain barrier competence in remote brain areas in ischemic stroke rats at the chronic stage. J Comp Neurol 2015; 522:3120-37. [PMID: 24610730 DOI: 10.1002/cne.23582] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/28/2014] [Accepted: 03/05/2014] [Indexed: 12/14/2022]
Abstract
Stroke is a life-threatening disease leading to long-term disability in stroke survivors. Cerebral functional insufficiency in chronic stroke might be due to pathological changes in brain areas remote from the initial ischemic lesion, i.e., diaschisis. Previously, we showed that the damaged blood-brain barrier (BBB) was involved in subacute diaschisis. The present study investigated BBB competence in chronic diaschisis by using a transient middle cerebral artery occlusion (tMCAO) rat model. Our results demonstrated significant BBB damage mostly in the ipsilateral striatum and motor cortex in rats at 30 days after tMCAO. The BBB alterations were also determined in the contralateral hemisphere via ultrastructural and immunohistochemical analyses. Major BBB pathological changes in contralateral remote striatum and motor cortex areas included 1) vacuolated endothelial cells containing large autophagosomes, 2) degenerated pericytes displaying mitochondria with cristae disruption, 3) degenerated astrocytes and perivascular edema, 4) Evans blue extravasation, and 5) appearance of parenchymal astrogliosis. Discrete analyses of striatal and motor cortex areas revealed significantly higher autophagosome accumulation in capillaries of ventral striatum and astrogliosis in dorsal striatum in both cerebral hemispheres. These widespread microvascular alterations in ipsilateral and contralateral brain hemispheres suggest persistent and/or continued BBB damage in chronic ischemia. The pathological changes in remote brain areas likely indicate chronic ischemic diaschisis, which should be considered in the development of treatment strategies for stroke.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612; Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612; Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612; Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612
| | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Li Q, Yang J. Improvement of Regional Cerebral Blood Flow After Stimulation of the Nucl. Fastigius in Cerebral Infarction Patients. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9526-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
59
|
Wang T, Zhai L, Guo Y, Pei H, Zhang M. Picroside II has a neuroprotective effect by inhibiting ERK1/2 activation after cerebral ischemic injury in rats. Clin Exp Pharmacol Physiol 2015; 42:930-939. [PMID: 26175147 DOI: 10.1111/1440-1681.12445] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/01/2015] [Accepted: 06/19/2015] [Indexed: 12/11/2022]
Abstract
In the study, the neuroprotective effect and underlying mechanism of picroside II were explored, and its involvement in the ERK1/2 signal pathway after cerebral ischemia injury in rats. A monofilament thread was inserted to generate middle cerebral artery occlusion (MCAO) in 100 Wistar rats that were administered an intraperitoneal injection of picroside II (20 mg/kg). The neurobehavioural function of rats was evaluated using a modified neurological severity score (mNSS) test. The cerebral infarct volume (CIV) was measured using tetrazolium chloride (TTC) staining. The morphology and ultra-structure of the nerve cells in the cortex were observed using hematoxylin and eosin (HE) staining and transmission electron microscopy (TEM), respectively. The apoptotic cells were counted using the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The expression of extracellular signal-regulated kinase 1/2 (pERK1/2) in the cortex was determined using immunohistochemistry and Western blot analysis. Neurological dysfunction was observed in all rats with MCAO. In both the model and lipopolysaccharide (LPS) groups, the CIV increased, the neuronal damage in the cortex was more severe, and the number of apoptotic cells and the pERK1/2 expression significantly increased compared with the control group (P < 0.05). In treatment and U0126 groups, the neurological function was improved, the CIV decreased, the neuronal damage in the cortex was attenuated, and the number of apoptotic cells and the pERK1/2 expression significantly decreased compared with the model group (P < 0.05). No significant differences in these indices were observed between model and LPS groups or treatment and U0126 groups (P > 0.05). The results suggest that activation of ERK1/2 in cerebral ischaemia induces neuronal apoptosis and picroside II may reduce neuronal apoptosis to confer protection against cerebral ischemic injury by inhibiting ERK1/2 activation.
Collapse
Affiliation(s)
- Tingting Wang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.,Taishan Scholars Construction Project Excellent Innovative Team of Shandong Province, Qingdao, China
| | - Li Zhai
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.,Taishan Scholars Construction Project Excellent Innovative Team of Shandong Province, Qingdao, China
| | - Yunliang Guo
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.,Taishan Scholars Construction Project Excellent Innovative Team of Shandong Province, Qingdao, China
| | - Haitao Pei
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.,Taishan Scholars Construction Project Excellent Innovative Team of Shandong Province, Qingdao, China
| | - Meizeng Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.,Taishan Scholars Construction Project Excellent Innovative Team of Shandong Province, Qingdao, China
| |
Collapse
|
60
|
Zuo XL, Deng HL, Wu P, Xu E. Do different reperfusion methods affect the outcomes of stroke induced by MCAO in adult rats? Int J Neurosci 2015; 126:850-5. [PMID: 26268737 DOI: 10.3109/00207454.2015.1074903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
There are two patterns of ischemia/reperfusion (I/R) models used in rat middle cerebral artery occlusion (MCAO) I/R models, which differ in the use of unilateral or bilateral carotid artery reperfusion. The primary difference between the two patterns of I/R models is the complexity of the surgery procedure. However, researchers in this field have no idea whether there are any differences in outcomes of these two methods. In this study, we investigated the effects of the two methods on neurological deficits, infarct volume, blood-brain barrier (BBB) integrity and brain derived neurotrophic factor (BDNF) expression. Through evaluating the current way of bilateral common carotid artery reperfusion, we tried to find whether it could be replaced by an easier way. We found that there were no statistical significant differences between the different methods in infarct volume, neurological deficits, BBB integrity, and the level of BDNF (P > 0.05). These data demonstrated that different methods did not affect the neurological deficits, infarct volume, BBB integrity, and the BDNF protein level, which provides reference when we use an experimental stroke. These results suggest that the two methods have similar capability for inducing cerebral I/R injury and can be interchanged.
Collapse
Affiliation(s)
- Xia-Lin Zuo
- a 1 Institute of Neurosciences and the Second Affiliated Hospital , Guangzhou Medical University , Guangzhou , P.R. China
| | - Hou-Liang Deng
- b 2 Center for Drug Research and Development, zhujiang Hospital , Southern Medical University , Guangzhou , P.R. China
| | - Ping Wu
- b 2 Center for Drug Research and Development, zhujiang Hospital , Southern Medical University , Guangzhou , P.R. China
| | - En Xu
- a 1 Institute of Neurosciences and the Second Affiliated Hospital , Guangzhou Medical University , Guangzhou , P.R. China
| |
Collapse
|
61
|
Picroside II Inhibits the MEK-ERK1/2-COX2 Signal Pathway to Prevent Cerebral Ischemic Injury in Rats. J Mol Neurosci 2015; 57:335-51. [PMID: 26240040 DOI: 10.1007/s12031-015-0623-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/14/2015] [Indexed: 10/23/2022]
Abstract
The objective of this study is to explore the neuroprotective effect and mechanism of picroside II on ERK1/2-COX2 signal transduction pathway after cerebral ischemic injury in rats. Focal cerebral ischemic models were established by inserting monofilament threads into the middle cerebral artery in 200 Wistar rats. Twenty four rats were randomly selected into control group, while the other rats were randomly divided into six groups: model group, picroside group, lipopolysaccharide (LPS) with picroside group, U0126 with picroside group, LPS group, and U0126 group with each group containing three subgroups with ischemia at 6, 12, and 24 h. Neurobehavioral function in the rats was evaluated by modified neurological severity score points (mNSS) test; structure of neurons was observed using hematoxylin-eosin (HE) staining; apoptotic cells were counted using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay; expressions of phosphorylated mitogen/extracellular signal-regulated kinase kinas1/2 (pMEK1/2), phosphorylated extracellular signal-regulated protein kinase1/2 (pERK1/2), and cyclooxygenase (COX2) in the cortex were determined using immunohistochemistry (IHC) and Western blot (WB); and real-time PCR was used to determine the level of COX2 mRNA. The neurological behavioral malfunction appeared in all rats with middle cerebral artery occlusion (MCAO). In the model group, neuron damage was extensive, while the neurobehavioral function score, apoptotic cell index, expression of pMEK1/2, pERK1/2, and COX2 and the level of COX2 mRNA increased significantly when compared to the control group. The peak COX2 mRNA level was in ischemia 12 h, prior to the peak in COX2 protein expression. In the picroside and U0126 groups, the neurological behavioral function was improved, and the number of apoptotic cells and the expression of pMEK1/2, pERK1/2, and COX2 decreased significantly when compared to the model group. In the LPS with picroside group, at ischemia 6 h neuron damage was extensive, and pMEK1/2, pERK1/2, and COX2 expression were much higher than in the model group. But at ischemia 12 and 24 h, the expression of pMEK1/2, pERK1/2, and COX2 decreased slightly, and the neurobehavioral function also improved slightly. In LPS group, neuron damage was extensive, pMEK1/2, pERK1/2, and COX2 expression was still at a high level, and COX2 mRNA peak arrived at ischemic 12 h. Picroside II downregulates COX2 expression after MCAO by inhibiting MEK-ERK1/2 in rats to protect neurons from apoptosis and inflammation.
Collapse
|
62
|
Abstract
The treatment for ischemic stroke is one of the most challenging problems and the therapeutic effect remains unsatisfied due to the poor permeation of drugs across the blood brain barrier (BBB). In this study, HAIYPRH (T7), a peptide that targeted to transferrin receptor (TfR) can mediate the transport of nanocarriers across the BBB, was conjugated to liposomes for ischemic stroke targeting treatment of a novel neuroprotectant (ZL006). T7-conjugated PEGylated liposomes (T7-P-LPs) loaded with ZL006 (T7-P-LPs/ZL006) were showed satisfactory vesicle size and size distribution. Furthermore, the cellular uptake results showed that T7 modification increased liposomes uptake by the brain capillary endothelial cells (BCECs) and little cytotoxicity of liposomes with or without ZL006 was observed. The in vivo biodistribution and near-infrared fluorescence imaging evidenced that T7 modification rendered liposomes significantly enhanced the transport of liposomes across the BBB. The pharmacodynamic study suggested that, T7-P-LPs/ZL006 exhibited reduced infarct volume and ameliorated neurological deficit compared with unmodified liposomes or free ZL006. T7-P-LPs/ZL006 could be targeted to brain and displayed remarkable neuroprotective effects. They could be used as a potential targeted drug delivery system of ischemic stroke treatment.
Collapse
|
63
|
Walberer M, Rueger MA. The macrosphere model-an embolic stroke model for studying the pathophysiology of focal cerebral ischemia in a translational approach. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207251 DOI: 10.3978/j.issn.2305-5839.2015.04.02] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The main challenge of stroke research is to translate promising experimental findings from the bench to the bedside. Many suggestions have been made how to achieve this goal, identifying the need for appropriate experimental animal models as one key issue. We here discuss the macrosphere model of focal cerebral ischemia in the rat, which closely resembles the pathophysiology of human stroke both in its acute and chronic phase. Key pathophysiological processes such as brain edema, cortical spreading depolarizations (CSD), neuroinflammation, and stem cell-mediated regeneration are observed in this stroke model, following characteristic temporo-spatial patterns. Non-invasive in vivo imaging allows studying the macrosphere model from the very onset of ischemia up to late remodeling processes in an intraindividual and longitudinal fashion. Such a design of pre-clinical stroke studies provides the basis for a successful translation into the clinic.
Collapse
Affiliation(s)
- Maureen Walberer
- 1 Department of Neurology, University Hospital of Cologne, Cologne, Germany ; 2 Max-Planck-Institute for Metabolism Research, Cologne, Germany ; 3 Animal Welfare Office, University of Cologne, Germany
| | - Maria Adele Rueger
- 1 Department of Neurology, University Hospital of Cologne, Cologne, Germany ; 2 Max-Planck-Institute for Metabolism Research, Cologne, Germany ; 3 Animal Welfare Office, University of Cologne, Germany
| |
Collapse
|
64
|
Sun M, Deng B, Zhao X, Gao C, Yang L, Zhao H, Yu D, Zhang F, Xu L, Chen L, Sun X. Isoflurane preconditioning provides neuroprotection against stroke by regulating the expression of the TLR4 signalling pathway to alleviate microglial activation. Sci Rep 2015; 5:11445. [PMID: 26086415 PMCID: PMC4471883 DOI: 10.1038/srep11445] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/20/2015] [Indexed: 01/05/2023] Open
Abstract
Excessive microglial activation often contributes to inflammation-mediated neurotoxicity in the ischemic penumbra during the acute stage of ischemic stroke. Toll-like receptor 4 (TLR4) has been reported to induce microglial activation via the NF-κB pathway. Isoflurane preconditioning (IP) can provide neuroprotection and inhibit microglial activation. In this study, we investigated the roles of the TLR4 signalling pathway in IP to exert neuroprotection following ischemic stroke in vivo and in vitro. The results showed that 2% IP alleviated neurological deficits, reduced the infarct volume, attenuated apoptosis and weakened microglial activation in the ischemic penumbra. Furthermore, IP down-regulated the expression of HSP 60, TLR4 and MyD88 and up-regulated inhibitor of IκB-α expression compared with I/R group in vivo. In vitro, 2% IP and a specific inhibitor of TLR4, CLI-095, down-regulated the expression of TLR4, MyD88, IL-1β, TNF-α and Bax, and up-regulated IκB-α and Bcl-2 expression compared with OGD group. Moreover, IP and CLI-095 attenuated microglial activation-induced neuronal apoptosis, and overexpression of the TLR4 gene reversed the neuroprotective effects of IP. In conclusion, IP provided neuroprotection by regulating TLR4 expression directly, alleviating microglial activation and neuroinflammation. Thus, inhibiting the activation of microglial activation via TLR4 may be a new avenue for stroke treatment.
Collapse
Affiliation(s)
- Meiyan Sun
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Bin Deng
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.,State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoyong Zhao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.,Department of Anesthesiology, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Changjun Gao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Lu Yang
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Hui Zhao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Daihua Yu
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Feng Zhang
- Department of Medical Administration, Lintong Sanatorium of PLA Lanzhou Military District, Lintong, Xi'an, 710600, China
| | - Lixian Xu
- State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Lei Chen
- Department of Gynaecology and Obstetrics, Nave General Hospital, Beijing, 100059, China
| | - Xude Sun
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| |
Collapse
|
65
|
Eckert A, Huang L, Gonzalez R, Kim HS, Hamblin MH, Lee JP. Bystander Effect Fuels Human Induced Pluripotent Stem Cell-Derived Neural Stem Cells to Quickly Attenuate Early Stage Neurological Deficits After Stroke. Stem Cells Transl Med 2015; 4:841-51. [PMID: 26025980 DOI: 10.5966/sctm.2014-0184] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 04/06/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED : Present therapies for stroke rest with tissue plasminogen activator (tPA), the sole licensed antithrombotic on the market; however, tPA's effectiveness is limited in that the drug not only must be administered less than 3-5 hours after stroke but often exacerbates blood-brain barrier (BBB) leakage and increases hemorrhagic incidence. A potentially promising therapy for stroke is transplantation of human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs). To date, the effects of iPSCs on injuries that take place during early stage ischemic stroke have not been well studied. Consequently, we engrafted iPSC-NSCs into the ipsilesional hippocampus, a natural niche of NSCs, at 24 hours after stroke (prior to secondary BBB opening and when inflammatory signature is abundant). At 48 hours after stroke (24 hours after transplant), hiPSC-NSCs had migrated to the stroke lesion and quickly improved neurological function. Transplanted mice showed reduced expression of proinflammatory factors (tumor necrosis factor-α, interleukin 6 [IL-6], IL-1β, monocyte chemotactic protein 1, macrophage inflammatory protein 1α), microglial activation, and adhesion molecules (intercellular adhesion molecule 1, vascular cell adhesion molecule 1) and attenuated BBB damage. We are the first to report that engrafted hiPSC-NSCs rapidly improved neurological function (less than 24 hours after transplant). Rapid hiPSC-NSC therapeutic activity is mainly due to a bystander effect that elicits reduced inflammation and BBB damage. SIGNIFICANCE Clinically, cerebral vessel occlusion is rarely permanent because of spontaneous or thrombolytic therapy-mediated reperfusion. These results have clinical implications indicating a much extended therapeutic window for transplantation of human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs; 24 hours after stroke as opposed to the 5-hour window with tissue plasminogen activator [tPA]). In addition, there is potential for a synergistic effect by combining hiPSC-NSC transplantation with tPA to attenuate stroke's adverse effects.
Collapse
Affiliation(s)
- Auston Eckert
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Lei Huang
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Rodolfo Gonzalez
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Hye-Sun Kim
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Milton H Hamblin
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Jean-Pyo Lee
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| |
Collapse
|
66
|
Fukuta T, Ishii T, Asai T, Nakamura G, Takeuchi Y, Sato A, Agato Y, Shimizu K, Akai S, Fukumoto D, Harada N, Tsukada H, Kawaguchi AT, Oku N. Real-time trafficking of PEGylated liposomes in the rodent focal brain ischemia analyzed by positron emission tomography. Artif Organs 2015; 38:662-6. [PMID: 25295359 DOI: 10.1111/aor.12350] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Aliposomal drug delivery system was previously applied to ischemic brain model rats for the treatment of brain ischemia, and we observed that 100-nm-sized liposomes could extravasate and accumulate in the ischemic brain region even when cerebral blood flow was markedly reduced in permanent middle cerebral artery occlusion (p-MCAO) model rats. In the present study, we investigated the real-time cerebral distribution of polyethylene glycol (PEG)-modified liposomes (PEGliposomes) labeled with 1-[18F]fluoro-3,6-dioxatetracosane in p-MCAO rats by positron emission tomography (PET). [18F]-Labeled PEG-liposomes were intravenously injected into p-MCAO rats 1 h after the onset of occlusion, and then a PET scan was performed for 2 h. The PET scan showed that the signal intensity of [18F] gradually increased in the ischemic region despite the drastic reduction in cerebral perfusion, suggesting that PEG-liposomes had accumulated in and around the ischemic region. Therefore,drug delivery to the ischemic region by use of liposomes would be possible under ischemic conditions, and a liposomal drug delivery system could be a promising strategy for protecting the ischemic brain from damage before recovery from ischemia.
Collapse
|
67
|
Picroside II Inhibits Neuronal Apoptosis and Improves the Morphology and Structure of Brain Tissue following Cerebral Ischemic Injury in Rats. PLoS One 2015; 10:e0124099. [PMID: 25927985 PMCID: PMC4415915 DOI: 10.1371/journal.pone.0124099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 02/26/2015] [Indexed: 12/29/2022] Open
Abstract
This paper aimed to explore the protective effects of picroside II against the neuronal apoptosis and changes in morphology and structure that follow cerebral ischemic injury in rats. A focal cerebral ischemic model was established by inserting a monofilament thread to achieve middle cerebral artery occlusion (MCAO) in 60 Wistar rats, and intraperitoneal injections of picroside II (20 mg/kg) were administered. The neurobehavioral functions were evaluated with the modified neurological severity score (mNSS) test. The cerebral infarct volumes were measured with tetrazolium chloride (TTC) staining. The morphology and ultrastructure of the cortical brain tissues were observed with hematoxylin-eosin staining and transmission electron microscopy, respectively. The apoptotic cells were counted with terminal deoxynucleotidyl transferase dUTP nick-end labeling and flow cytometry, and pERK1/2 expression was determined by immunohistochemical assay and Western blot. The results indicated that neurological behavioral malfunctions and cerebral infarcts were present in the MCAO rats. In the model group, the damage to the structures of the neurons and the blood brain barrier (BBB) in the cortex was more severe, and the numbers of apoptotic cells, the early apoptotic ratio (EAR) and pERK1/2 expression were significantly increased in this group compared to the control group (P<0.05). In the treatment group, the neurological behavioral function and the morphology and ultrastructure of the neurons and the BBB were improved including the number of Mi increased and relative area of condensed chromosome and basement (BM) thickness descreased, and the cerebral infarct volume, the number of apoptotic cells, the EAR and pERK1/2 expression were significantly decreased compared to the model group (P<0.05). These results suggest that picroside II reduced apoptosis and improved the morphology and ultrastructure of the neurons and the BBB and that these effects resulted in the recovery of the neurobehavioral function of rats with cerebral ischemia.
Collapse
|
68
|
Neuroprotective effect of 6-paradol in focal cerebral ischemia involves the attenuation of neuroinflammatory responses in activated microglia. PLoS One 2015; 10:e0120203. [PMID: 25789481 PMCID: PMC4366308 DOI: 10.1371/journal.pone.0120203] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/20/2015] [Indexed: 12/27/2022] Open
Abstract
Paradols are non-pungent and biotransformed metabolites of shogaols and reduce inflammatory responses as well as oxidative stress as shogaols. Recently, shogaol has been noted to possess therapeutic potential against several central nervous system (CNS) disorders, including cerebral ischemia, by reducing neuroinflammation in microglia. Therefore, paradol could be used to improve neuroinflammation-associated CNS disorders. Here, we synthesized paradol derivatives (2- to 10-paradols). Through the initial screening for anti-inflammatory activities using lipopolysaccharide (LPS)-stimulated BV2 microglia, 6-paradol was chosen to be the most effective compound without cytotoxicity. Pretreatment with 6-paradol reduced neuroinflammatory responses in LPS-stimulated BV2 microglia by a concentration-dependent manner, which includes reduced NO production by inhibiting iNOS upregulation and lowered secretion of proinflammatory cytokines (IL-6 and TNF-α). To pursue whether the beneficial in vitro effects of 6-paradol leads towards in vivo therapeutic effects on transient focal cerebral ischemia characterized by neuroinflammation, we employed middle cerebral artery occlusion (MCAO)/reperfusion (M/R). Administration of 6-paradol immediately after reperfusion significantly reduced brain damage in M/R-challenged mice as assessed by brain infarction, neurological deficit, and neural cell survival and death. Furthermore, as observed in cultured microglia, 6-paradol administration markedly reduced neuroinflammation in M/R-challenged brains by attenuating microglial activation and reducing the number of cells expressing iNOS and TNF-α, both of which are known to be produced in microglia following M/R challenge. Collectively, this study provides evidences that 6-paradol effectively protects brain after cerebral ischemia, likely by attenuating neuroinflammation in microglia, suggesting it as a potential therapeutic agent to treat cerebral ischemia.
Collapse
|
69
|
Choi Y, Kang SG, Kam KY. Changes in the BDNF-immunopositive cell population of neocortical layers I and II/III after focal cerebral ischemia in rats. Brain Res 2015; 1605:76-82. [PMID: 25681548 DOI: 10.1016/j.brainres.2015.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 12/17/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family and is widely distributed in the central nervous system, including the cerebral cortex. BDNF plays an important role in normal neural development, survival of existing neurons, and activity-dependent neuroplasticity. BDNF can also be neuroprotective and evoke neurogenesis in certain pathological conditions, such as cerebral ischemia. Neocortical layer I is an important region that can integrate feedforward and feedback information from other cortical areas and subcortical regions. In addition, it has recently been proposed as a possible source of neuronal progenitor cells after ischemia. Therefore, we investigated changes in the BDNF-immunoreactive cell population of neocortical layers I and II/III after middle cerebral artery occlusion (MCAO)-induced cerebral ischemia in rats. In unaffected condition, the number of BDNF(+) cells in layer I was significantly less than in layer II/III in the cingulate cortex and in the motor and sensory areas. The increase in the number of BDNF(+) cells in layer I 8 days after MCAO was more remarkable than layer II/III, in all regions except the area of cingulate cortex farthest from the infarct core. Only BDNF(+)-Ox-42(+) cells showed a tendency to increase consistently toward the infarct core in both layers I and II/III, implying a major source of BDNF for response to ischemic injury. The present study suggests that some beneficial effects during recovery from ischemic injury, such as increased supportive microglia/macrophages, occur owing to a sensitive response of BDNF in layer I.
Collapse
Affiliation(s)
- Yongwon Choi
- Department of Rehabilitation Science, Inje University, Gimhae 621-749, Republic of Korea; U-Healthcare & Anti-aging Research Center, Inje University, Gimhae 621-749, Republic of Korea
| | - Sung Goo Kang
- Department of Biological Sciences, Institute of Basic Science, Inje University, Gimhae 621-749, Republic of Korea
| | - Kyung-Yoon Kam
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Republic of Korea; U-Healthcare & Anti-aging Research Center, Inje University, Gimhae 621-749, Republic of Korea.
| |
Collapse
|
70
|
Xu Y, Du S, Yu X, Han X, Hou J, Guo H. Human bone marrow mesenchymal stem cell transplantation attenuates axonal injury in stroke rats. Neural Regen Res 2015; 9:2053-8. [PMID: 25657721 PMCID: PMC4316468 DOI: 10.4103/1673-5374.147930] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2014] [Indexed: 01/01/2023] Open
Abstract
Previous studies have shown that transplantation of human bone marrow mesenchymal stem cells promotes neural functional recovery after stroke, but the neurorestorative mechanisms remain largely unknown. We hypothesized that functional recovery of myelinated axons may be one of underlying mechanisms. In this study, an ischemia/reperfusion rat model was established using the middle cerebral artery occlusion method. Rats were used to test the hypothesis that intravenous transplantation of human bone marrow mesenchymal stem cells through the femoral vein could exert neuroprotective effects against cerebral ischemia via a mechanism associated with the ability to attenuate axonal injury. The results of behavioral tests, infarction volume analysis and immunohistochemistry showed that cerebral ischemia caused severe damage to the myelin sheath and axons. After rats were intravenously transplanted with human bone marrow mesenchymal stem cells, the levels of axon and myelin sheath-related proteins, including microtubule-associated protein 2, myelin basic protein, and growth-associated protein 43, were elevated, infarct volume was decreased and neural function was improved in cerebral ischemic rats. These findings suggest that intravenously transplanted human bone marrow mesenchymal stem cells promote neural function. Possible mechanisms underlying these beneficial effects include resistance to demyelination after cerebral ischemia, prevention of axonal degeneration, and promotion of axonal regeneration.
Collapse
Affiliation(s)
- Yi Xu
- Department of Neurosurgery, General Hospital of Chinese PLA, Beijing, China
| | - Shiwei Du
- Department of Neurosurgery, General Hospital of Armed Police Forces, Beijing, China
| | - Xinguang Yu
- Department of Neurosurgery, General Hospital of Chinese PLA, Beijing, China
| | - Xiao Han
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Institute of Basic Medical Sciences, Beijing, China ; Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medcal Sciences of Xuyuan Hospital, Beijing, China
| | - Jincai Hou
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Institute of Basic Medical Sciences, Beijing, China ; Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medcal Sciences of Xuyuan Hospital, Beijing, China
| | - Hao Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Institute of Basic Medical Sciences, Beijing, China ; Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medcal Sciences of Xuyuan Hospital, Beijing, China
| |
Collapse
|
71
|
Nik Ramli NN, Omar N, Husin A, Ismail Z, Siran R. Preconditioning effect of (S)-3,5-dihydroxyphenylglycine on ischemic injury in middle cerebral artery occluded Sprague-Dawley rats. Neurosci Lett 2015; 588:137-41. [PMID: 25562631 DOI: 10.1016/j.neulet.2014.12.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/30/2014] [Accepted: 12/31/2014] [Indexed: 10/24/2022]
Abstract
Glutamate receptors are the integral cellular components associated with excitotoxicity mechanism induced by the ischemic cascade events. Therefore the glutamate receptors have become the major molecular targets of neuroprotective agents in stroke researches. Recent studies have demonstrated that a Group I metabotropic glutamate receptor agonist, (S)-3,5-dihydroxyphenylglycine ((S)-3,5-DHPG) preconditioning elicits neuroprotection in the hippocampal slice cultures exposed to toxic level of N-methyl-d-aspartate (NMDA). We further investigated the preconditioning effects of (S)-3,5-DHPG on acute ischemic stroke rats. One 10 or 100μM of (S)-3,5-DHPG was administered intrathecally to Sprague-Dawley adult male rats, 2h prior to induction of acute ischemic stroke by middle cerebral artery occlusion (MCAO). After 24h, neurological deficits were evaluated by modified stroke severity scores and grid-walking test. All rats were sacrificed and infarct volumes were determined by 2,3,5-triphenyltetrazolium chloride staining. The serum level of neuron-specific enolase (NSE) of each rat was analyzed by enzyme-linked immunosorbent assay (ELISA). One and 10μM of (S)-3,5-DHPG preconditioning in the stroke rats showed significant improvements in motor impairment (P<0.01), reduction in the infarct volume (P<0.01) and reduction in the NSE serum level (P<0.01) compared to the control stroke rats. We conclude that 1 and 10μM (S)-3,5-DHPG preconditioning induced protective effects against acute ischemic insult in vivo.
Collapse
Affiliation(s)
- Nik Nasihah Nik Ramli
- Institute of Medical Molecular Biotechnology, Universiti Teknologi MARA, 47000 Selangor, Malaysia
| | - Nursyazwani Omar
- Institute of Medical Molecular Biotechnology, Universiti Teknologi MARA, 47000 Selangor, Malaysia
| | - Andrean Husin
- Faculty of Dentistry, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia; Brain and Neuroscience Communities of Research, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Zalina Ismail
- Brain Research and Information Network, Centre for Neurocognitive Sciences, Universiti Sains Malaysia, 16150 Kelantan, Malaysia
| | - Rosfaiizah Siran
- Brain and Neuroscience Communities of Research, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia.
| |
Collapse
|
72
|
Zhang Q, Guo P, Wang J, Yang M, Kong L. Gender-specific metabolic responses in focal cerebral ischemia of rats and Huang-Lian-Jie-Du decoction treatment. RSC Adv 2015. [DOI: 10.1039/c5ra19934d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
1H NMR based metabolomics approach combined with biochemical, histological and immunohistochemistry observations was successfully applied to explore gender-specific metabolic differences in ischemic stroke and the protective effect of HLJDD.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- PR China
| | - Pingping Guo
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- PR China
| | - Junsong Wang
- Center for Molecular Metabolism
- Nanjing University of Science & Technology
- Nanjing 210094
- PR China
| | - Minghua Yang
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- PR China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- PR China
| |
Collapse
|
73
|
Balsara RD, Chapman SE, Sander IM, Donahue DL, Liepert L, Castellino FJ, Leevy WM. Non-invasive imaging and analysis of cerebral ischemia in living rats using positron emission tomography with 18F-FDG. J Vis Exp 2014:51495. [PMID: 25590998 PMCID: PMC4354491 DOI: 10.3791/51495] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Stroke is the third leading cause of death among Americans 65 years of age or older(1). The quality of life for patients who suffer from a stroke fails to return to normal in a large majority of patients(2), which is mainly due to current lack of clinical treatment for acute stroke. This necessitates understanding the physiological effects of cerebral ischemia on brain tissue over time and is a major area of active research. Towards this end, experimental progress has been made using rats as a preclinical model for stroke, particularly, using non-invasive methods such as (18)F-fluorodeoxyglucose (FDG) coupled with Positron Emission Tomography (PET) imaging(3,10,17). Here we present a strategy for inducing cerebral ischemia in rats by middle cerebral artery occlusion (MCAO) that mimics focal cerebral ischemia in humans, and imaging its effects over 24 hr using FDG-PET coupled with X-ray computed tomography (CT) with an Albira PET-CT instrument. A VOI template atlas was subsequently fused to the cerebral rat data to enable a unbiased analysis of the brain and its sub-regions(4). In addition, a method for 3D visualization of the FDG-PET-CT time course is presented. In summary, we present a detailed protocol for initiating, quantifying, and visualizing an induced ischemic stroke event in a living Sprague-Dawley rat in three dimensions using FDG-PET.
Collapse
Affiliation(s)
- Rashna D Balsara
- W. M. Keck Center for Transgene Research, University of Notre Dame; Department of Chemistry and Biochemistry, University of Notre Dame
| | - Sarah E Chapman
- Notre Dame Integrated Imaging Facility, University of Notre Dame
| | - Ian M Sander
- Department of Biological Sciences, University of Notre Dame
| | | | - Lucas Liepert
- Department of Biological Sciences, University of Notre Dame
| | - Francis J Castellino
- W. M. Keck Center for Transgene Research, University of Notre Dame; Department of Chemistry and Biochemistry, University of Notre Dame
| | - W Matthew Leevy
- Notre Dame Integrated Imaging Facility, University of Notre Dame; Department of Biological Sciences, University of Notre Dame; Harper Cancer Research Institute, University of Notre Dame;
| |
Collapse
|
74
|
Neurovascular protection by telmisartan via reducing neuroinflammation in stroke-resistant spontaneously hypertensive rat brain after ischemic stroke. J Stroke Cerebrovasc Dis 2014; 24:537-47. [PMID: 25534368 DOI: 10.1016/j.jstrokecerebrovasdis.2014.09.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 09/11/2014] [Accepted: 09/29/2014] [Indexed: 11/22/2022] Open
Abstract
Telmisartan is a highly lipid-soluble angiotensin receptor blocker (ARB), which improves insulin sensitivity and reduces triglyceride levels and, thus, is called metabo-sartan. We examined the effects of telmisartan on neurovascular unit (N-acetylglucosamine oligomer [NAGO], collagen IV, and glial fibrillary acidic protein [GFAP]) and neuroinflammation (matrix metalloproteinase-9 [MMP-9] and inflammasome) in brain of stroke-resistant spontaneously hypertensive rat (SHR-SR). At 12 weeks of age, SHR-SR received transient middle cerebral artery occlusion (tMCAO) for 90 minutes and were divided into the following 3 groups, that is, vehicle group, low-dose telmisartan group (.3 mg/kg/d), and high-dose telmisartan group (3 mg/kg/d, postoral). Immunohistologic analysis at ages 6, 12, and 18 months showed progressive decreases of NAGO-positive endothelium and collagen IV-positive basement membrane and progressive increases of MMP-9-positive neurons, GFAP-positive astrocytes, and NLRP3-positive inflammasome in the cerebral cortex of vehicle group. Low-dose telmisartan reduced such changes without lowering blood pressure (BP), and high-dose telmisartan further improved such changes with lowering BP. The present findings suggest that a persistent hypertension caused a long-lasting inflammation after tMCAO in SHR-SR, which accelerated neurovascular disruption and emergent inflammasome, and that telmisartan greatly reduced such inflammation and protected the neurovascular unit via its pleiotropic effects in living hypertensive rat brain after ischemic stroke.
Collapse
|
75
|
Taninishi H, Jung JY, Izutsu M, Wang Z, Sheng H, Warner DS. A blinded randomized assessment of laser Doppler flowmetry efficacy in standardizing outcome from intraluminal filament MCAO in the rat. J Neurosci Methods 2014; 241:111-20. [PMID: 25526908 DOI: 10.1016/j.jneumeth.2014.12.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/07/2014] [Accepted: 12/09/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND Laser Doppler flowmetry (LDF) is widely used for estimating cerebral blood flow changes during intraluminal middle cerebral artery occlusion (MCAO). No investigation has systematically examined LDF efficacy in standardizing outcome. We examined MCAO histologic and behavioral outcome as a function of LDF measurement. MATERIALS AND METHODS Rats were subjected to 90min MCAO by 4 surgeons having different levels of MCAO surgical experience. LDF was measured in all rats during ischemia. By random assignment, LDF values were (Assisted) or were not (Blinded) made available to each surgeon during MCAO (n=12-17 per group). Neurologic and histologic outcomes were measured 7 days post-MCAO. A second study examined LDF effects on 1-day post-MCAO outcome. RESULTS Pooled across surgeons, intra-ischemic %LDF change (P=0.12), neurologic scores (Assisted vs. Blinded=14±6 vs. 13±7, P=0.61, mean±standard deviation) and cerebral infarct volume (162±63mm(3)vs. 143±86mm(3), P=0.24) were not different between groups. Only for one surgeon (novice) did LDF use alter infarct volume (145±28mm(3)vs. 98±61mm(3), P=0.03). LDF use decreased infarct volume coefficient of variation (COV) by 35% (P=0.02), but had no effect on neurologic score COV. COMPARISON WITH EXISTING METHODS We compared intraluminal MCAO outcome as a function of LDF use. CONCLUSIONS LDF measurement altered neither neurologic nor histologic MCAO outcome. LDF did not decrease neurologic deficit COV, but did decrease infarct volume COV. LDF may allow use of fewer animals if infarct volume is the primary dependent variable, but is unlikely to impact requisite sample sizes if neurologic function is of primary interest.
Collapse
Affiliation(s)
- Hideki Taninishi
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC, United States; Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Jin Yong Jung
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC, United States; Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Miwa Izutsu
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC, United States; Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Zhengfeng Wang
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC, United States; Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Huaxin Sheng
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC, United States; Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - David S Warner
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC, United States; Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States; Department of Surgery, Duke University Medical Center, Durham, NC, United States; Department of Neurobiology, Duke University Medical Center, Durham, NC, United States.
| |
Collapse
|
76
|
Zhang H, Song LC, Jia CH, Lu YL. Effects of ATP sensitive potassium channel opener on the mRNA and protein expressions of caspase-12 after cerebral ischemia-reperfusion in rats. Neurosci Bull 2014; 24:7-12. [PMID: 18273070 DOI: 10.1007/s12264-008-1227-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE To investigate effects of K(ATP) opener on the expressions of caspase-12 mRNA and protein, and to explore the role of endoplasmic reticulum (ER) stress pathway in the mechanism of K(ATP) opener protecting against neuronal apoptosis after cerebral ischemia-reperfusion. METHODS Two hundred rats were randomly divided into four groups: sham operation group, ischemia-reperfusion group, K(ATP) opener group, and K(ATP) blocker group. The middle cerebral artery occlusion (MCAO) model was established by intraluminal suture occlusion method; neuronal apoptosis was detected by TUNEL staining. The mRNA and protein expressions of caspase-12 were detected by semi-quantitative RT-PCR and immunohistochemical staining, respectively. RESULTS In ischemia-reperfusion group, K(ATP) opener group and K(ATP) blocker group, the number of apoptotic cells and the mRNA and protein expressions of caspase-12 gradually increased following cerebral reperfusion, and reached the peak at 24 h. In K(ATP) opener group, the number of apoptotic cells was significantly less than that in ischemia-reperfusion group and K(ATP) blocker group at 12 h, 24 h, 48 h and 72 h (P< 0.05 or P< 0.01); while the mRNA and protein levels of caspase-12 were significantly less than those in ischemia-reperfusion group and K(ATP) blocker group at all times (P< 0.05 or P< 0.01). There were no differences between the ischemia-reperfusion group and K(ATP) blocker group at each time (P> 0.05). CONCLUSION K(ATP) opener may protect neurons from apoptosis following the cerebral ischemia-reperfusion by inhibiting ER stress pathway.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Neurology, the Affiliated Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | | | | | | |
Collapse
|
77
|
Matrix Metalloproteinase-8 is a Novel Pathogenetic Factor in Focal Cerebral Ischemia. Mol Neurobiol 2014; 53:231-239. [PMID: 25421209 DOI: 10.1007/s12035-014-8996-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 11/11/2014] [Indexed: 12/20/2022]
Abstract
The neutrophil collagenase matrix metalloproteinase-8 (MMP8) is a recently identified member of MMPs that have important roles in various inflammation-related disorders. Previously, we identified MMP8 as a new neuroinflammatory mediator in activated microglia by regulating TNF-α productivity. Here, we present evidence that MMP8 is a critical factor for brain damage in transient focal cerebral ischemia by modulating neuroinflammation likely microglial activation and TNF-α production. Biochemical analyses showed upregulation of MMP8 expression at mRNA and protein levels in transient middle cerebral artery occlusion/reperfusion (M/R)-challenged brains. Furthermore, double immunolabeling showed that MMP8 expression was upregulated in the activated microglia of M/R-challenged brains. Assessment of infarct volume, neurological score, and survival/death of neural cells revealed that administration of an MMP8 inhibitor (M8I) immediately after reperfusion reduced brain damage. Histological analyses showed that microglial activation and TNF-α expression in ischemic conditions was abrogated by exposure to M8I, as demonstrated in our previous study using cultured microglia. These outcomes from a pharmacological approach were reaffirmed by a genetic approach using a lentiviral system. Intracerebroventricular microinjection of MMP8-specific shRNA lentivirus reduced the extent of ischemia-induced brain damage, as assessed by infarct volume, neurological score, microglial activation, and TNF-α expression. These results suggest a novel pathogenetic role of MMP8 and implicate modulation of its activity as a tractable strategy for therapies against cerebral ischemia.
Collapse
|
78
|
Huang L, Wong S, Snyder EY, Hamblin MH, Lee JP. Human neural stem cells rapidly ameliorate symptomatic inflammation in early-stage ischemic-reperfusion cerebral injury. Stem Cell Res Ther 2014; 5:129. [PMID: 25418536 PMCID: PMC4445985 DOI: 10.1186/scrt519] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 11/04/2014] [Indexed: 01/03/2023] Open
Abstract
Introduction Clinically, a good deal of injury from stroke results from ischemic-reperfusion. There is a loss of cerebral parenchyma and its associated cells, disruption of neuronal connections, compromise of the blood-brain barrier, and inflammation. We tested whether exogenously engrafted human neural stem cells could migrate rapidly and extensively to damaged regions, following transplantation into a neurogenic site where migration cues are already underway during stroke onset, then counteract a number of these pathological processes. Methods One day post-injury, we injected human neural stem cells (hNSCs) into the ipsilesional hippocampus of a mouse model of stroke with middle cerebral artery occlusion to induce focal ischemia followed by reperfusion (MCAO/R). The time frame for hNSC transplantation corresponded to upregulation of endogenous proinflammatory cytokines. We examined the effect of hNSC transplantation on pathological processes and behavioral dysfunction 48 hours post-injury. Results Twenty-four hours after transplantation, engrafted hNSCs had migrated extensively to the lesion, and infarct volume was reduced relative to MCAO/R controls. The behavioral deficits seen in MCAO/R controls were also significantly improved. Given this rapid response, we hypothesized that the mechanisms underlying therapeutic activity were anti-inflammatory rather than due to cell replacement. In support of this idea, in hNSC-transplanted mice we observed reduced microglial activation, decreased expression of proinflammatory factors (tumor necrosis factor-α, interleukin (IL)-6, IL-1β, monocyte chemotactic protein-1, macrophage inflammatory protein-1α) and adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1), and amelioration of blood-brain barrier damage. Conclusions While long-term effects of engrafted hNSCs on the amelioration of ischemic stroke-induced behavioral dysfunction in a rodent model have been reported, our study is the first to show rapid, beneficial impacts on behavioral function (within 24 hours) upon early delivery of hNSCs into the hippocampus. Electronic supplementary material The online version of this article (doi:10.1186/scrt519) contains supplementary material, which is available to authorized users.
Collapse
|
79
|
Selvamani A, Williams MH, Miranda RC, Sohrabji F. Circulating miRNA profiles provide a biomarker for severity of stroke outcomes associated with age and sex in a rat model. Clin Sci (Lond) 2014; 127:77-89. [PMID: 24428837 PMCID: PMC4386587 DOI: 10.1042/cs20130565] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Small non-coding RNA [miRNA (microRNA)] found in the circulation have been used successfully as biomarkers and mechanistic targets for chronic and acute disease. The present study investigated the impact of age and sex on miRNA expression following ischaemic stroke in an animal model. Adult (6 month) and middle-aged (11-12 months) female and male rats were subject to MCAo (middle cerebral artery occlusion) using ET-1 (endothelin-1). Circulating miRNAs were analysed in blood samples at 2 and 5 days post-stroke, and brain miRNAs were analysed at 5 days post-stroke. Although stroke-associated infarction was observed in all groups, infarct volume and sensory-motor deficits were significantly reduced in adult females compared with middle-aged females, adult males or middle-aged males. At 2 days post-stroke, 21 circulating miRNAs were differentially regulated and PCA (principal component analysis) confirmed that most of the variance was due to age. At 5 days post-stroke, 78 circulating miRNAs exhibited significantly different regulation, and most of the variance was associated with sex. A small cohort (five) of miRNAs, miR-15a, miR-19b, miR-32 miR-136 and miR-199a-3p, were found to be highly expressed exclusively in adult females compared with middle-aged females, adult males and middle-aged males. Predicted gene targets for these five miRNAs analysed for KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways revealed that the top ten KEGG pathways were related to growth factor signalling, cell structure and PI3K (phosphoinositide 3-kinase)/Akt and mTOR (mammalian target of rapamycin) signalling. Overall, the pattern of circulating miRNA expression suggests an early influence of age in stroke pathology, with a later emergence of sex as a factor for stroke severity.
Collapse
Affiliation(s)
- Amutha Selvamani
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, U.S.A
| | - Madison H. Williams
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, U.S.A
| | - Rajesh C. Miranda
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, U.S.A
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, U.S.A
| |
Collapse
|
80
|
Choi JI, Kim SD, Kim SH, Lim DJ, Ha SK. Semi-quantitative analyses of hippocampal heat shock protein-70 expression based on the duration of ischemia and the volume of cerebral infarction in mice. J Korean Neurosurg Soc 2014; 55:307-12. [PMID: 25237425 PMCID: PMC4166325 DOI: 10.3340/jkns.2014.55.6.307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/19/2014] [Accepted: 06/11/2014] [Indexed: 11/27/2022] Open
Abstract
Objective We investigated the expression of hippocampal heat shock protein 70 (HSP-70) infarction volume after different durations of experimental ischemic stroke in mice. Methods Focal cerebral ischemia was induced in mice by occluding the middle cerebral artery with the modified intraluminal filament technique. Twenty-four hours after ischemia induction, both hippocampi were extracted for HSP-70 protein analyses. Slices from each hemisphere were stained with 2,3,5-triphenyltetrazolium chloride (2%), and infarction volumes were calculated. HSP-70 levels were evaluated using western blot and enzyme-linked immunosorbent assay (ELISA). HSP-70 subtype (hsp70.1, hspa1a, hspa1b) mRNA levels in the hippocampus were measured using reverse transcription-polymerase chain reaction (RT-PCR). Results Cerebral infarctions were found ipsilateral to the occlusion in 10 mice exposed to transient ischemia (5 each in the 30-min and 60-min occlusion groups), whereas no focal infarctions were noted in any of the sham mice. The average infarct volumes of the 2 ischemic groups were 22.28±7.31 mm3 [30-min group±standard deviation (SD)] and 38.06±9.53 mm3 (60-min group±SD). Western blot analyses and ELISA showed that HSP-70 in hippocampal tissues increased in the infarction groups than in the sham group. However, differences in HSP-70 levels between the 2 infarction groups were statistically insignificant. Moreover, RT-PCR results demonstrated no relationship between the mRNA expression of HSP-70 subtypes and occlusion time or infarction volume. Conclusion Our results indicated no significant difference in HSP-70 expression between the 30- and 60-min occlusion groups despite the statistical difference in infarction volumes. Furthermore, HSP-70 subtype mRNA expression was independent of both occlusion duration and cerebral infarction volume.
Collapse
Affiliation(s)
- Jong-Il Choi
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Sang-Dae Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Se-Hoon Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Dong-Jun Lim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Sung-Kon Ha
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| |
Collapse
|
81
|
Wu SK, Yang MT, Kang KH, Liou HC, Lu DH, Fu WM, Lin WL. Targeted delivery of erythropoietin by transcranial focused ultrasound for neuroprotection against ischemia/reperfusion-induced neuronal injury: a long-term and short-term study. PLoS One 2014; 9:e90107. [PMID: 24587228 PMCID: PMC3938648 DOI: 10.1371/journal.pone.0090107] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 01/29/2014] [Indexed: 11/20/2022] Open
Abstract
Erythropoietin (EPO) is a neuroprotective agent against cerebral ischemia/reperfusion (I/R)-induced brain injury. However, its crossing of blood-brain barrier is limited. Focused ultrasound (FUS) sonication with microbubbles (MBs) can effectively open blood-brain barrier to boost the vascular permeability. In this study, we investigated the effects of MBs/FUS on extending the therapeutic time window of EPO and its neuroprotective effects in both acute and chronic phases. Male Wistar rats were firstly subjected to two common carotid arteries and right middle cerebral artery occlusion (three vessels occlusion, 3VO) for 50 min, and then the rats were treated with hEPO (human recombinant EPO, 5000 IU/kg) with or without MBs/FUS at 5 h after occlusion/reperfusion. Acute phase investigation (I/R, I/R+MBs/FUS, I/R+hEPO, and I/R+hEPO+MBs/FUS) was performed 24 h after I/R; chronic tests including cylinder test and gait analysis were performed one month after I/R. The experimental results showed that MBs/FUS significantly increased the cerebral content of EPO by bettering vascular permeability. In acute phase, both significant improvement of neurological score and reduction of infarct volume were found in the I/R+hEPO+MBs/FUS group, as compared with I/R and I/R+hEPO groups. In chronic phase, long-term behavioral recovery and neuronal loss in brain cortex after I/R injury was significantly improved in the I/R+hEPO+MBs/FUS group. This study indicates that hEPO administration with MBs/FUS sonication even at 5 h after occlusion/reperfusion can produce a significant neuroprotection.
Collapse
Affiliation(s)
- Sheng-Kai Wu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Ming-Tao Yang
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Kai-Hsiang Kang
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Houng-Chi Liou
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dai-Hua Lu
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Mei Fu
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail: (WL); (WF)
| | - Win-Li Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
- Division of Medical Engineering Research, National Health Research Institutes, Miaoli, Taiwan
- * E-mail: (WL); (WF)
| |
Collapse
|
82
|
A new method for detecting cerebral hemorrhage in rabbits by magnetic inductive phase shift. Biosens Bioelectron 2014; 52:374-8. [DOI: 10.1016/j.bios.2013.09.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 09/05/2013] [Accepted: 09/09/2013] [Indexed: 11/20/2022]
|
83
|
Protective effect of serofendic acid, administered intravenously, on cerebral ischemia-reperfusion injury in rats. Brain Res 2013; 1532:99-105. [DOI: 10.1016/j.brainres.2013.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 07/26/2013] [Accepted: 08/05/2013] [Indexed: 01/09/2023]
|
84
|
Zhu S, Li Y, Lu H, Li H, Tong S. Imaging the early cerebral blood flow changes in rat middle cerebral artery occlusion stroke model. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2012:2655-8. [PMID: 23366471 DOI: 10.1109/embc.2012.6346510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Intraluminal middle cerebral artery occlusion (MCAO) model in rats has been widely used to mimic human ischemic stroke and serves as an indispensable tool in the stroke research field. One limitation of this model is its high variability in infarct volume. The cerebral blood flow (CBF) information after cerebrovascular occlusion may reflect the availability of collateral circulation, which serves as a key factor for brain infarct volume. Laser speckle contrast imaging (LSCI) is a valuable tool for full-field imaging of CBF with high spatial and temporal resolution. In this paper, we investigated the spatio-temporal changes of CBF in rat MCAO stroke model using our self-developed real-time LSCI system. CBF images of adult male Sprague Dawley rats (n=13) were recorded before surgery, during first 1.5 hours after surgery, and 24 hours after stroke. We compared the CBF changes of different functional vessels during this period. In the ipsilateral hemisphere, CBF of veins and arteries both decreased as expected, while CBF of veins increased after occlusion in the contralateral hemisphere. Moreover, we found a linear correlation between early-stage CBF after occlusion and brain infarct volume, which can be utilized for surgery guidance to improve the uniformity of rat MCAO stroke models.
Collapse
Affiliation(s)
- Shuping Zhu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | | | | | | | | |
Collapse
|
85
|
Prieto R, Carceller F, Roda JM, Avendaño C. The intraluminal thread model revisited: rat strain differences in local cerebral blood flow. Neurol Res 2013; 27:47-52. [PMID: 15829158 DOI: 10.1179/016164105x18214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
There are major differences in the outcome of focal cerebral ischemia between rat strains. This study aimed to investigate whether inter-strain differences exist in the local cerebral blood flow, changes produced by intraluminal middle cerebral artery occlusion (MCAO). Fifty-four male Long-Evans, Sprague-Dawley or Wistar rats were subjected to 60 minutes of transient MCAO, carried out with a silicone-coated 4-0 nylon monofilament. Local cerebral blood flow was monitored on the parietal cortex with laser-Doppler flowmetry. The decrease in local cerebral blood flow during the first 20 minutes of occlusion was significantly greater in Long-Evans (74.6 +/- 8.9%) than in Sprague-Dawley (50.7 +/- 26.8%) and Wistar (49.0 +/- 21.9%) rats. Strain differences in local cerebral blood flow reduction were maintained, or increased, throughout the occlusion period. These results show that the intraluminal model of MCAO produces a more pronounced, persistent and stable local cerebral blood flow reduction, with less interanimal variability, in Long-Evans than in Sprague-Dawley or Wistar rats.
Collapse
Affiliation(s)
- Ruth Prieto
- Service of Neurosurgery, University Hospital La Paz, Madrid, Spain.
| | | | | | | |
Collapse
|
86
|
Hung YC, Chou YS, Chang CH, Lin HW, Chen HY, Chen TY, Tai SH, Lee EJ. Early reperfusion improves the recovery of contralateral electrophysiological diaschisis following focal cerebral ischemia in rats. Neurol Res 2013; 32:828-34. [DOI: 10.1179/016164109x12581096870032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
87
|
Zhu H, Qian Z, He F, Liu M, Pan L, Zhang Q, Tang Y. Novel pharmacokinetic studies of the Chinese formula Huang-Lian-Jie-Du-Tang in MCAO rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:767-774. [PMID: 23628154 DOI: 10.1016/j.phymed.2012.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 11/06/2012] [Accepted: 11/21/2012] [Indexed: 06/02/2023]
Abstract
Our previous studies showed that after oral administration of an Huang-Lian-Jie-Du-Tang (HLJDT) decoction, there is a higher concentration of the pure components, berberine, baicalin and gardenoside in the plasma of Middle cerebral artery occlusion (MCAO) rats than in sham-operated rats, The aim of the present study was to determine whether these components could be reliably measured in MCAO rat tissues. First, the plasma concentration-time profiles of berberine, palmatine, baicalin, baicalein and gardenoside were characterised in MCAO rats after oral administration of the aqueous extract of HLJDT. Subsequently, liver, lung and kidney tissues were obtained from sudden death MCAO rats in the absorption phase (0.25 h), the distribution phase (1.0 h) and the elimination phase (8.0 h) after administration of the HLJDT aqueous extract. An HPLC method was developed and validated for the determination of the distribution characteristics of berberine, palmatine, baicalin, baicalein and gardenoside simultaneously from the above-mentioned rat tissues. The results indicated that berberine, palmatine, baicalin and baicalein distributed rapidly and accumulated at high levels in the lung, while gardenoside distributed widely in the lung and the kidney. To the best of our knowledge, this is the first report to describe the distribution of the active ingredients derived from HLJDT in MCAO rat tissues. The tissue distribution results provide a biopharmaceutical basis for the design of the clinic application of HLJDT in cerebrovascular disease.
Collapse
Affiliation(s)
- Huaxu Zhu
- Separation Engineering of Chinese Traditional Medicine Compound, Nanjing University of Chinese Medicine, Nanjing 210029, China.
| | | | | | | | | | | | | |
Collapse
|
88
|
Wang W, Kang J, Li H, Su J, Wu J, Xu Y, Yu H, Xiang X, Yi H, Lu Y, Sun L. Regulation of endoplasmic reticulum stress in rat cortex by p62/ZIP through the Keap1-Nrf2-ARE signalling pathway after transient focal cerebral ischaemia. Brain Inj 2013; 27:924-33. [PMID: 23782269 DOI: 10.3109/02699052.2013.793397] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PRIMARY OBJECTIVE p62/ZIP as the autophagy receptor can transport the misfolded proteins to a macroautophagy-lysosome system for degradation and also create a positive feedback loop between p62/ZIP and Nrf2. However, the role of p62/ZIP on cerebral ischaemia is unclear. The aim of this study was to evaluate the role of p62/ZIP in the regulation of endoplasmic reticulum(ER) stress induced by cerebral ischaemia/reperfusion. RESEARCH DESIGN Different ischemic periods were designed by transient middle cerebral artery occlusion (tMCAO) using the suture method. METHODS AND PROCEDURES At 24 hours after reperfusion, the ischaemic brain tissue was studied histologically and biochemically for autophagic, ER stress and Keap1-Nrf2-ARE signalling pathway markers. MAIN OUTCOMES AND RESULTS Prolongation of ischaemia significantly increased the cortical injury observed in rats and was associated with a gradual increase in the protein expression of ubiquitin-aggregates, Grp78, GADD153/CHOP and p62/ZIP. Autophagy marker Atg12-Atg5 and LC3-PE increased and then decreased. Moreover, p62/ZIP mRNA expression increased and then decreased and was consistent with Nrf2 activation. CONCLUSIONS p62/ZIP not only plays a key role in scavenging protein aggregates during autophagy, but it may also be involved in preventing oxidative injury and alleviating ER stress through the Keap1-Nrf2-ARE signalling pathway during cerebral ischaemia/reperfusion injury.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, 126 Xinmin Street, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Garbuzova-Davis S, Rodrigues MCO, Hernandez-Ontiveros DG, Tajiri N, Frisina-Deyo A, Boffeli SM, Abraham JV, Pabon M, Wagner A, Ishikawa H, Shinozuka K, Haller E, Sanberg PR, Kaneko Y, Borlongan CV. Blood-brain barrier alterations provide evidence of subacute diaschisis in an ischemic stroke rat model. PLoS One 2013; 8:e63553. [PMID: 23675488 PMCID: PMC3651135 DOI: 10.1371/journal.pone.0063553] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 04/02/2013] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Comprehensive stroke studies reveal diaschisis, a loss of function due to pathological deficits in brain areas remote from initial ischemic lesion. However, blood-brain barrier (BBB) competence in subacute diaschisis is uncertain. The present study investigated subacute diaschisis in a focal ischemic stroke rat model. Specific focuses were BBB integrity and related pathogenic processes in contralateral brain areas. METHODOLOGY/PRINCIPAL FINDINGS In ipsilateral hemisphere 7 days after transient middle cerebral artery occlusion (tMCAO), significant BBB alterations characterized by large Evans Blue (EB) parenchymal extravasation, autophagosome accumulation, increased reactive astrocytes and activated microglia, demyelinization, and neuronal damage were detected in the striatum, motor and somatosensory cortices. Vascular damage identified by ultrastuctural and immunohistochemical analyses also occurred in the contralateral hemisphere. In contralateral striatum and motor cortex, major ultrastructural BBB changes included: swollen and vacuolated endothelial cells containing numerous autophagosomes, pericyte degeneration, and perivascular edema. Additionally, prominent EB extravasation, increased endothelial autophagosome formation, rampant astrogliosis, activated microglia, widespread neuronal pyknosis and decreased myelin were observed in contralateral striatum, and motor and somatosensory cortices. CONCLUSIONS/SIGNIFICANCE These results demonstrate focal ischemic stroke-induced pathological disturbances in ipsilateral, as well as in contralateral brain areas, which were shown to be closely associated with BBB breakdown in remote brain microvessels and endothelial autophagosome accumulation. This microvascular damage in subacute phase likely revealed ischemic diaschisis and should be considered in development of treatment strategies for stroke.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Ishii T, Fukuta T, Agato Y, Oyama D, Yasuda N, Shimizu K, Kawaguchi AT, Asai T, Oku N. Nanoparticles accumulate in ischemic core and penumbra region even when cerebral perfusion is reduced. Biochem Biophys Res Commun 2013; 430:1201-5. [DOI: 10.1016/j.bbrc.2012.12.080] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 12/14/2012] [Indexed: 01/12/2023]
|
91
|
Dalkara T, Arsava EM. Can restoring incomplete microcirculatory reperfusion improve stroke outcome after thrombolysis? J Cereb Blood Flow Metab 2012; 32:2091-9. [PMID: 23047270 PMCID: PMC3519416 DOI: 10.1038/jcbfm.2012.139] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 08/31/2012] [Accepted: 09/03/2012] [Indexed: 12/27/2022]
Abstract
Substantial experimental data and recent clinical evidence suggesting that tissue reperfusion is a better predictor of outcome after thrombolysis than recanalization necessitate that patency of microcirculation after recanalization should be reevaluated. If indeed microcirculatory blood flow cannot be sufficiently reinstituted despite complete recanalization as commonly observed in coronary circulation, it may be one of the factors contributing to low efficacy of thrombolysis in stroke. Although microvascular no-reflow is considered an irreversible process that prevents tissue recovery from injury, emerging evidence suggests that it might be reversed with pharmacological agents administered early during recanalization. Therefore, therapeutic approaches aiming at reducing microvascular obstructions may improve success rate of recanalization therapies. Importantly, promoting oxygen delivery to the tissue, where entrapped erythrocytes cannot circulate in capillaries, with ongoing serum flow may improve survival of the underreperfused tissue. Altogether, these developments bring about the exciting possibility that benefit of reperfusion therapies can be further improved by restoring microcirculatory function because survival in the penumbra critically depends on adequate blood supply. Here, we review the available evidence suggesting presence of an 'incomplete microcirculatory reperfusion' (IMR) after focal cerebral ischemia and discuss potential means that may help investigate IMR in stroke patients after recanalization therapies despite technical limitations.
Collapse
Affiliation(s)
- Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| | | |
Collapse
|
92
|
Pabbidi MR, Juncos J, Juncos L, Renic M, Tullos HJ, Lazar J, Jacob HJ, Harder DR, Roman RJ. Identification of a region of rat chromosome 1 that impairs the myogenic response and autoregulation of cerebral blood flow in fawn-hooded hypertensive rats. Am J Physiol Heart Circ Physiol 2012; 304:H311-7. [PMID: 23144316 DOI: 10.1152/ajpheart.00622.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study examined the effects of transfer of a 2.4-Mbp region of rat chromosome 1 (RNO1) from Brown Norway (BN) into fawn-hooded hypertensive (FHH) rats on autoregulation (AR) of cerebral blood flow (CBF) and the myogenic response of middle cerebral arteries (MCAs). AR of CBF was poor in FHH and FHH.1(BN) AR(-) congenic strains that excluded the critical 2.4-Mbp region. In contrast, AR was restored in FHH.1(BN) AR(+) congenic strains that included this region. The diameter of MCAs of FHH rats increased from 140 ± 14 to 157 ± 18 μm when transmural pressure was increased from 40 to 140 mmHg, but it decreased from 137 ± 5 to 94 ± 7 μm in FHH.1(BN) AR(+) congenic strains. Transient occlusion of MCAs reduced CBF by 80% in all strains. However, the hyperemic response following ischemia was significantly greater in FHH and AR(-) rats than that seen in AR(+) congenic strains (AR(-), 173 ± 11% vs. AR(+), 124 ± 5%). Infarct size and edema formation were also significantly greater in an AR(-) strain (38.6 ± 2.6 and 12.1 ± 2%) than in AR(+) congenic strains (27.6 ± 1.8 and 6.5 ± 0.9%). These results indicate that there is a gene in the 2.4-Mbp region of RNO1 that alters the development of myogenic tone in cerebral arteries. Transfer of this region from BN to FHH rats restores AR of CBF and vascular reactivity and reduces cerebral injury after transient occlusion and reperfusion of the MCA.
Collapse
|
93
|
Kam KY, Jalin AMA, Choi YW, Kaengkan P, Park SW, Kim YH, Kang SG. Ziprasidone attenuates brain injury after focal cerebral ischemia induced by middle cerebral artery occlusion in rats. Prog Neuropsychopharmacol Biol Psychiatry 2012; 39:69-74. [PMID: 22627197 DOI: 10.1016/j.pnpbp.2012.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 04/29/2012] [Accepted: 05/14/2012] [Indexed: 11/29/2022]
Abstract
Ziprasidone is an atypical antipsychotic drug used for the treatment of schizophrenia. Recent studies have reported that atypical antipsychotics have neuroprotective effects against brain injury. In the present study, the effect of ziprasidone on ischemic brain injury was investigated. Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in rats. All the animals experienced ischemia for 1h and then underwent reperfusion. The infarct size induced by MCAO was significantly reduced in the animals that received acute treatment with 5mg/kg ziprasidone and subchronic treatment with 2.5mg/kg ziprasidone for 7 days compared with that in the vehicle-treated animals. The acute treatment with ziprasidone significantly improved neurological functions, as measured by the modified neurological severity score, in a dose-dependent manner. The subchronic treatment produced more rapid recovery from functional deficits than the vehicle treatment. The immunohistochemical investigation revealed that the subchronic treatment prevented severe loss of neuronal marker intensity and attenuated the increased in microglial marker intensity in the infarcted cortical area. These results suggest that ziprasidone has neuroprotective effects in a rat model of ischemic stroke and provide new insight for its clinical applications.
Collapse
Affiliation(s)
- Kyung-Yoon Kam
- Department of Occupational Therapy, Inje University, Gimhae 621-749, South Korea
| | | | | | | | | | | | | |
Collapse
|
94
|
Gu Q, Gu H, Lu X, Lu F, Shao Y, Zhang S. The influence of deep hypothermic global brain ischemia on EEG in a new rat model. J Card Surg 2012; 27:612-7. [PMID: 22978840 DOI: 10.1111/j.1540-8191.2012.01498.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Neurological complications following deep hypothermic circulatory arrest (DHCA) occur between t 4% ≈ 25%. However, the cerebral injury mechanisms are still not well understood due to a lack of a practical and simple animal model. We aimed to establish a rodent deep hypothermic global brain ischemia (DHGBI) model, which can be used to elucidate these mechanisms in future studies. DESIGN 30 Sprague-Dawley rats were divided randomly into three groups: the carotid occlusion DHGBI group, the internal carotid shunt DHGBI group, and the sham operation group. We validated the model in terms of electroencephalogram (EEG) and regional cerebral blood flow (rCBF). All rats were sacrificed for analysis of brain moisture capacity after 24 hours. RESULTS In the internal carotid shunt DHGBI group the EEG activity was suppressed to "flat-line" and the relative power of the α and θ frequency bands was decreased (p < 0.05). However, in the carotid occlusion DHGBI group we only observed the relative power of the α frequency band depressed (p < 0.05). The rCBF was significantly decreased in all groups. In the internal carotid shunt DHGBI group the rCBF was significantly reduced to 4.27 ± 2.75%, and was lower than the other two groups (p < 0.05). The result of brain moisture capacity was consistent with the EEG and rCBF observations. CONCLUSIONS The current study presents a novel cerebral recovery model of DHCA in the rat. This experimental model may be suitable to further elucidate the mechanisms associated with adverse cerebral outcomes after DHCA and to investigate potential neuroprotective strategies.
Collapse
Affiliation(s)
- Qun Gu
- Jiangsu Province Hospital, First Affiliated Hospital with Nanjing Medical University, Department of Cardiothoracic Surgery, Nanjing, China
| | | | | | | | | | | |
Collapse
|
95
|
Ishii T, Asai T, Fukuta T, Oyama D, Yasuda N, Agato Y, Shimizu K, Minamino T, Oku N. A single injection of liposomal asialo-erythropoietin improves motor function deficit caused by cerebral ischemia/reperfusion. Int J Pharm 2012; 439:269-74. [PMID: 22989979 DOI: 10.1016/j.ijpharm.2012.09.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/28/2012] [Accepted: 09/09/2012] [Indexed: 11/25/2022]
Abstract
Modification of the liposomal surface with a targeting molecule is a promising approach for the targeted delivery of therapeutics. Asialo-erythropoietin (AEPO) is a potent tool for targeting an ischemic region by binding to the EPO receptors on neuronal cells. Additionally, it shows a strong cytoprotective effect against programed cell death. Hence, AEPO-modified liposomes appear likely to have both a neuronal-targeting character and a neuroprotective effect on cerebral ischemic injury. In this study, we assessed the targeting ability of AEPO-modified PEGylated liposomes (AEPO-liposomes) to ischemic region and their improvement effect on neurological deficits induced by ischemia/reperfusion (I/R) in transient middle cerebral artery occlusion (t-MCAO) rats. Immunohistological analysis showed that the AEPO-liposomes given immediately after reperfusion extravasated into the ischemic region and attached strongly to neuronal cells. Also, neuronal nuclei (NeuN) staining was clearly visible only in the AEPO-liposome-treated group, suggesting that AEPO-liposomes protected neuronal cells from ischemia/reperfusion-induced damage. Moreover, a single administration of low-dose AEPO-liposomes significantly improved the neurological deficit compared to vehicle and free-AEPO treatment at 7 days after injection. In conclusion, AEPO-liposomes have clear potential as a neuroprotectant after stroke and as a DDS device targeting ischemic regions.
Collapse
Affiliation(s)
- Takayuki Ishii
- Department of Medical Biochemistry and Global COE, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Shizuoka 422-8526, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Lee NY, Park HYL, Park CK, Ahn MD. Analysis of systemic endothelin-1, matrix metalloproteinase-9, macrophage chemoattractant protein-1, and high-sensitivity C-reactive protein in normal-tension glaucoma. Curr Eye Res 2012; 37:1121-6. [PMID: 22966842 DOI: 10.3109/02713683.2012.725798] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To investigate the roles of vascular dysregulation and inflammation in normal-tension glaucoma (NTG), we determined the plasma levels of endothelin-1 (ET-1), matrix metalloproteinase-9 (MMP-9), macrophage chemoattractant protein-1 (MCP-1), and high-sensitivity C-reactive protein (hs-CRP). MATERIALS AND METHODS Forty-five patients with NTG and age-matched 35 healthy controls were enrolled in this study. Blood samples from all subjects were assayed for ET-1, MMP-9, MCP-1, and hs-CRP concentrations and other systemic factors. RESULTS There were no significant differences in hemoglobin, hematocrit, RBC count, WBC count, platelet count, fasting glucose, HbA1c, total cholesterol, triglyceride, LDL, and HDL between the NTG and control groups. The systemic levels of ET-1 and MCP-1 were significantly higher in the NTG group than in the control group (p = 0.05 and 0.02, respectively). The MMP-9 and hs-CRP levels were not significantly different between the NTG and control groups. CONCLUSIONS After excluding patients with cardiovascular and other systemic diseases, plasma ET-1 and MCP-1 levels were elevated in patients with NTG. The MMP-9 and hs-CRP levels were not significantly different in NTG. Increased ET-1 and MCP-1 levels suggest that ischemia/inflammation may play a role in the pathogenesis of NTG.
Collapse
Affiliation(s)
- Na Young Lee
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | |
Collapse
|
97
|
Increase of 20-HETE synthase after brain ischemia in rats revealed by PET study with 11C-labeled 20-HETE synthase-specific inhibitor. J Cereb Blood Flow Metab 2012; 32:1737-46. [PMID: 22669478 PMCID: PMC3434634 DOI: 10.1038/jcbfm.2012.68] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
20-Hydroxyeicosatetraenoic acid (20-HETE), an arachidonic acid metabolite known to be produced after cerebral ischemia, has been implicated in ischemic and reperfusion injury by mediating vasoconstriction. To develop a positron emission tomography (PET) probe for 20-HETE synthase imaging, which might be useful for monitoring vasoconstrictive processes in patients with brain ischemia, we synthesized a (11)C-labeled specific 20-HETE synthase inhibitor, N'(4-dimethylaminohexyloxy)phenyl imidazole ([(11)C]TROA). Autoradiographic study showed that [(11)C]TROA has high-specific binding in the kidney and liver consistent with the previously reported distribution of 20-HETE synthase. Using transient middle cerebral artery occlusion in rats, PET study showed significant increases in the binding of [(11)C]TROA in the ipsilateral hemisphere of rat brains after 7 and 10 days, which was blocked by co-injection of excess amounts of TROA (10 mg/kg). The increased [(11)C]TROA binding on the ipsilateral side returned to basal levels within 14 days. In addition, quantitative real-time PCR revealed that increased expression of 20-HETE synthase was only shown on the ipsilateral side on day 7. These results indicate that [(11)C]TROA might be a useful PET probe for imaging of 20-HETE synthase in patients with cerebral ischemia.
Collapse
|
98
|
Martín A, Macé E, Boisgard R, Montaldo G, Thézé B, Tanter M, Tavitian B. Imaging of perfusion, angiogenesis, and tissue elasticity after stroke. J Cereb Blood Flow Metab 2012; 32:1496-507. [PMID: 22491156 PMCID: PMC3421095 DOI: 10.1038/jcbfm.2012.49] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Blood flow interruption in a cerebral artery causes brain ischemia and induces dramatic changes of perfusion and metabolism in the corresponding territory. We performed in parallel positron emission tomography (PET) with [(15)O]H(2)O, single photon emission computed tomography (SPECT) with [(99m)Tc]hexamethylpropylene-amino-oxime ([(99m)Tc]HMPAO) and ultrasonic ultrafast shear wave imaging (SWI) during, immediately after, and 1, 2, 4, and 7 days after middle cerebral artery occlusion (MCAO) in rats. Positron emission tomography and SPECT showed initial hypoperfusion followed by recovery at immediate reperfusion, hypoperfusion at day 1, and hyperperfusion at days 4 to 7. Hyperperfusion interested the whole brain, including nonischemic areas. Immunohistochemical analysis indicated active angiogenesis at days 2 to 7, strongly suggestive that hyperperfusion was supported by an increase in microvessel density in both brain hemispheres after ischemia. The SWI detected elastic changes of cerebral tissue in the ischemic area as early as day 1 after MCAO appearing as a softening of cerebral tissue whose local internal elasticity decreased continuously from day 1 to 7. Taken together, these results suggest that hyperperfusion after cerebral ischemia is due to formation of neovessels, and indicate that brain softening is an early and continuous process. The SWI is a promising novel imaging method for monitoring the evolution of cerebral ischemia over time in animals.
Collapse
Affiliation(s)
- Abraham Martín
- Inserm U1023, Université Paris Sud, CEA, DSV, I2BM, Orsay, France
| | | | | | | | | | | | | |
Collapse
|
99
|
Jung SY, Kim HJ, Lee JY, Cho JS, Lee YS, Jin CB. Neuroprotective Effects of Quercetin 3-O-Methyl Ether, Quercetin and (±)-Dihydroquercetin in a Rat Model of Transient Focal Cerebral Ischemia. B KOREAN CHEM SOC 2012. [DOI: 10.5012/bkcs.2012.33.7.2443] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
100
|
Li C, Feng JJ, Wu YP, Zhang GY. Cerebral ischemia-reperfusion induces GAPDH S-nitrosylation and nuclear translocation. BIOCHEMISTRY (MOSCOW) 2012; 77:671-8. [DOI: 10.1134/s0006297912060156] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|