51
|
Wu L, Su Z, Zha L, Zhu Z, Liu W, Sun Y, Yu P, Wang Y, Zhang G, Zhang Z. Tetramethylpyrazine Nitrone Reduces Oxidative Stress to Alleviate Cerebral Vasospasm in Experimental Subarachnoid Hemorrhage Models. Neuromolecular Med 2019; 21:262-274. [PMID: 31134485 DOI: 10.1007/s12017-019-08543-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 05/10/2019] [Indexed: 12/13/2022]
Abstract
Cerebral vasospasm is one of the deleterious complications after subarachnoid hemorrhage (SAH), leading to delayed cerebral ischemia and permanent neurological deficits or even death. Free radicals and oxidative stress are considered as crucial causes contributing to cerebral vasospasm and brain damage after SAH. Tetramethylpyrazine nitrone (TBN), a derivative of the clinically used anti-stroke drug tetramethylpyrazine armed with a powerful free radical scavenging nitrone moiety, has been reported to prevent brain damage from ischemic stroke. The present study aimed to investigate the effects of TBN on vasospasm and brain damage after SAH. Two experimental SAH models were used, a rat model by endovascular perforation and a rabbit model by intracisternal injection of autologous blood. The effects of TBN on SAH were evaluated assessing basilar artery spasm, neuronal apoptosis, and neurological deficits. TBN treatment significantly attenuated vasospasm, improved neurological behavior functions and reduced the number of apoptotic neurons in both the SAH rats and rabbits. Mechanistically, TBN suppressed the increase in 3-nitrotyrosine and 8-hydroxy-2-deoxyguanosine immuno-positive cells in the cortex of SAH rat brain. Western blot analyses indicated that TBN effectively reversed the altered expression of Bcl-2, Bax and cytochrome C, and up-regulated nuclear factor erythroid-derived 2-like 2 (Nrf2) and hemeoxygenase-1 (HO-1) protein expressions. In the in vitro studies, TBN inhibited H2O2-induced bEnd.3 cell apoptosis and reduced ROS generation. Additionally, TBN alleviated the contraction of rat basilar artery rings induced by H2O2 ex vivo. In conclusion, TBN ameliorated SAH-induced cerebral vasospasm and neuronal damage. These effects of TBN may be attributed to its anti-oxidative stress effect and up-regulation of Nrf2/HO-1.
Collapse
Affiliation(s)
- Liangmiao Wu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Zhiyang Su
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Ling Zha
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Zeyu Zhu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Wei Liu
- Foshan Magpie Pharmaceuticals Co., LTD, Foshan, Guangdong Province, China
| | - Yewei Sun
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Pei Yu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Yuqiang Wang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China
| | - Gaoxiao Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China.
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Huangpu Road, Guangzhou, China.
| |
Collapse
|
52
|
Apelin-13 attenuates early brain injury following subarachnoid hemorrhage via suppressing neuronal apoptosis through the GLP-1R/PI3K/Akt signaling. Biochem Biophys Res Commun 2019; 513:105-111. [DOI: 10.1016/j.bbrc.2019.03.151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 03/23/2019] [Indexed: 11/20/2022]
|
53
|
Yang X, Zhang Y, Liu E, Sun L, Zhang Y, Chen C, Wang A, Yan J. The effects of dihydroxyphenyl lactic acid on alleviating blood-brain barrier injury following subarachnoid hemorrhage in rats. Neurosci Lett 2019; 704:189-194. [PMID: 30980845 DOI: 10.1016/j.neulet.2019.04.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND In this study, the protective effects of 3,4-dihydroxyphenyl lactic acid (DLA) on blood-brain barrier (BBB) injury following subarachnoid hemorrhage (SAH) has been explored. METHODS Male Sprague-Dawley rats (weight 300-350 g) were used to establish the SAH model using the endovascular perforation method. The animals were randomly divided into four groups: sham (n = 40), SAH (n = 46), SAH + vehicle (n = 44), and SAH + DLA (n = 40) treatment groups. At 1 h after SAH, either DLA (10 mg/kg) or normal saline (vehicle) was administered by femoral vein injection. The effects of DLA on mortality, neurological function, brain water content, and BBB were observed. Additionally, immunohistochemistry and western blot techniques were applied to investigate the mechanism of action of DLA. RESULTS We found that the administration of DLA (10 mg/kg) following SAH could improve neurological functions, reduce brain water content, and maintain BBB integrity. The expression of pro-inflammatory and pro-apoptotic factors such as toll-like receptor 4 (TLR4), NF-κB (p-p65), tumor necrosis factor-α, p-p38 MAPK, p-p53, and caspase-3 were significantly increased after SAH. These same factors were markedly attenuated following treatment with DLA. CONCLUSIONS These findings showed that DLA can alleviate BBB injury following SAH through its anti-inflammatory and anti-apoptotic effects via suppression of TLR4 and its downstream NF-κB and p38 MAPK pathways.
Collapse
Affiliation(s)
- Xiaomei Yang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yixuan Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - E Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Linlin Sun
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yan Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Chunhua Chen
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Aibo Wang
- Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Junhao Yan
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, 100191, China.
| |
Collapse
|
54
|
Shen F, Jiang L, Han F, Degos V, Chen S, Su H. Increased Inflammatory Response in Old Mice is Associated with More Severe Neuronal Injury at the Acute Stage of Ischemic Stroke. Aging Dis 2019; 10:12-22. [PMID: 30705764 PMCID: PMC6345332 DOI: 10.14336/ad.2018.0205] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
Stroke occurs mostly in patients with advanced age. Elderly patients have a less favorable prognosis compared with young adult patients. To understand the underlying mechanisms, we tested our hypothesis that an increased inflammatory response to acute ischemic injury in old stroke mice leads to more severe brain damage and behavioral dysfunction. An ischemic stroke model was created in 2- and 12-month-old C57BL/6 mice through permanent occlusion of the left distal middle cerebral artery (dMCAO). Infarct/atrophy volumes were quantified by staining the brain sections with Cresyl Violet. Sensorimotor function was assessed using the corner test and adhesive removal test. Quantification of CD68+ cells in the peri-infarct region was performed at 1, 3 and 14 days after dMCAO. Interleukin-6 (IL-6), interleukin-1 β (IL-1β) and vascular endothelial growth factor (VEGF) levels in the ischemic brain tissue were measured using ELISA. Western blot was used to determine the expression levels of tight junction proteins, claudin-5 and zonula occludens (ZO)-1. Blood-brain barrier permeability was measured by Evans blue (EB) extravasation. Gelatinase B (MMP-9, type IV collagenase) was measured by gel zymography. Compared to 2-month-old mice, 12-month-old mice had more severe behavioral deficits at both the acute and chronic stages of stroke. Compared with the 2-month-old mice, 12-month-old mice had larger infarct/atrophy volumes at 1 and 14 days after dMCAO, higher levels of IL-6 and IL-1β, higher MMP9 activity, and lower levels of claudin-5 and ZO-1 at 1 and 3 days after dMCAO. 12-month-old mice also had more CD68+ cells in the peri-infarct region at 1, 3 and 14 days after dMCAO and more EB leakage at 3 days after dMCAO. A higher inflammatory response at the acute stage of ischemic stroke in old mice is associated with more severe neuronal injury and long-term behavioral dysfunction.
Collapse
Affiliation(s)
- Fanxia Shen
- 1Department of Neurology & Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Lidan Jiang
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Frank Han
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Vincent Degos
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Shengdi Chen
- 1Department of Neurology & Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Su
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
55
|
Wang X, Li S, Ma J, Wang C, Chen A, Xin Z, Zhang J. Effect of Gastrodin on Early Brain Injury and Neurological Outcome After Subarachnoid Hemorrhage in Rats. Neurosci Bull 2019; 35:461-470. [PMID: 30673960 DOI: 10.1007/s12264-018-00333-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/10/2018] [Indexed: 12/01/2022] Open
Abstract
Gastrodin is a phenolic glycoside that has been demonstrated to provide neuroprotection in preclinical models of central nervous system disease, but its effect in subarachnoid hemorrhage (SAH) remains unclear. In this study, we showed that intraperitoneal administration of gastrodin (100 mg/kg per day) significantly attenuated the SAH-induced neurological deficit, brain edema, and increased blood-brain barrier permeability in rats. Meanwhile, gastrodin treatment significantly reduced the SAH-induced elevation of glutamate concentration in the cerebrospinal fluid and the intracellular Ca2+ overload. Moreover, gastrodin suppressed the SAH-induced microglial activation, astrocyte activation, and neuronal apoptosis. Mechanistically, gastrodin significantly reduced the oxidative stress and inflammatory response, up-regulated the expression of nuclear factor erythroid 2-related factor 2, heme oxygenase-1, phospho-Akt and B-cell lymphoma 2, and down-regulated the expression of BCL2-associated X protein and cleaved caspase-3. Our results suggested that the administration of gastrodin provides neuroprotection against early brain injury after experimental SAH.
Collapse
Affiliation(s)
- Xinzhi Wang
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Linqing, 252601, China
| | - Shuyue Li
- Department of Internal Medicine, The Second People's Hospital of Liaocheng, Linqing, 252601, China
| | - Jinbang Ma
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Linqing, 252601, China
| | - Chuangang Wang
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Linqing, 252601, China
| | - Anzhong Chen
- Department of Rehabilitation Medicine, The Second People's Hospital of Liaocheng, Linqing, 252601, China
| | - Zhenxue Xin
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Linqing, 252601, China.
| | - Jianjun Zhang
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Linqing, 252601, China.
| |
Collapse
|
56
|
Wang W, Han P, Xie R, Yang M, Zhang C, Mi Q, Sun B, Zhang Z. TAT-mGluR1 Attenuation of Neuronal Apoptosis through Prevention of MGluR1α Truncation after Experimental Subarachnoid Hemorrhage. ACS Chem Neurosci 2019; 10:746-756. [PMID: 30339347 DOI: 10.1021/acschemneuro.8b00531] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excessive glutamate-mediated overactivation of metabotropic glutamate receptor 1 (mGluR1) plays a leading role in neuronal apoptosis following subarachnoid hemorrhage (SAH). TAT-mGluR1, a fusion peptide consisting of a peptide spanning the calpain cleavage site of mGluR1α and the trans-activating regulatory protein (TAT) of HIV, effectively blocks mGluR1α truncation and protects neurons against excitotoxic damage. This study investigated the effects of TAT-mGluR1 on neuronal apoptosis in the rat SAH model. Here, we report that SAH caused activation of calpain and truncation of mGluR1α; intraperitoneally administered TAT-mGluR1 did not affect calpain activity, while it blocked truncation of mGluR1α after SAH. Intraperitoneally administered FITC-labeled TAT-mGluR1 was colocalized with mGluR1α in thecortex after SAH. Furthermore, TAT-mGluR1 significantly improved the neurological deficit, increased p-PI3K, p-Akt, and p-GSK3β, downregulated Bax, upregulated Bcl-2, and reduced cortical apoptosis in the basal cortex at 24 h after SAH. These findings indicated that TAT-mGluR1 acted against SAH-induced cell apoptosis through preventing mGluR1α truncation.
Collapse
Affiliation(s)
- Weiqi Wang
- Medical College of Qingdao University, Qingdao 266021, Shandong, People’s Republic of China
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| | - Ping Han
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| | - Rongxia Xie
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
- Department of Neurology, Central Hospital of Xinwen Mining Group, Taian 271000, People’s Republic of China
| | - Mingfeng Yang
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| | - Cheng Zhang
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| | - Qiongjie Mi
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| | - Baoliang Sun
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| | - Zongyong Zhang
- Key Lab of Cerebral Microcirculation at the Universities of Shandong, Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian 271016, Shandong, People’s Republic of China
| |
Collapse
|
57
|
Liu ZW, Zhao JJ, Pang HG, Song JN. Vascular endothelial growth factor A promotes platelet adhesion to collagen IV and causes early brain injury after subarachnoid hemorrhage. Neural Regen Res 2019; 14:1726-1733. [PMID: 31169190 PMCID: PMC6585561 DOI: 10.4103/1673-5374.257530] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The role of vascular endothelial growth factor A in platelet adhesion in cerebral microvessels in the early stage of subarachnoid hemorrhage remains unclear. In this study, the endovascular puncture method was used to produce a rat model of subarachnoid hemorrhage. Then, 30 minutes later, vascular endothelial growth factor A antagonist anti-vascular endothelial growth factor receptor 2 antibody, 10 μg, was injected into the right ventricle. Immunohistochemistry and western blot assay were used to assess expression of vascular endothelial growth factor A, occludin and claudin-5. Immunohistochemical double labeling was conducted to examine co-expression of GP Ia-II integrin and type IV collagen. TUNEL was used to detect apoptosis in the hippocampus. Neurological score was used to assess behavioral performance. After subarachnoid hemorrhage, the expression of vascular endothelial growth factor A increased in the hippocampus, while occludin and claudin-5 expression levels decreased. Co-expression of GP Ia-II integrin and type IV collagen and the number of apoptotic cells increased, whereas behavioral performance was markedly impaired. After treatment with anti-vascular endothelial growth factor receptor 2 antibody, occludin and claudin-5 expression recovered, while co-expression of GP Ia-II integrin and type IV collagen and the number of apoptotic cells decreased. Furthermore, behavioral performance improved notably. Our findings suggest that increased vascular endothelial growth factor A levels promote platelet adhesion and contribute to early brain injury after subarachnoid hemorrhage. This study was approved by the Biomedical Ethics Committee, Medical College of Xi’an Jiaotong University, China in December 2015.
Collapse
Affiliation(s)
- Zun-Wei Liu
- Department of Renal Transplantation, Nephropathy Hospital, the First Affiliated Hospital, Medical College of Xi'an Jiaotong University; Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jun-Jie Zhao
- Department of Neurosurgery, the First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hong-Gang Pang
- The First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jin-Ning Song
- Department of Neurosurgery, the First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
58
|
Petridis AK, Kamp MA, Cornelius JF, Beez T, Beseoglu K, Turowski B, Steiger HJ. Aneurysmal Subarachnoid Hemorrhage. DEUTSCHES ARZTEBLATT INTERNATIONAL 2018; 114:226-236. [PMID: 28434443 DOI: 10.3238/arztebl.2017.0226] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 08/29/2016] [Accepted: 11/28/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND Aneurysmal subarachnoid hemorrhage (SAH) is associated with a mortality of more than 30%. Only about 30% of patients with SAB recover sufficiently to return to independent living. METHODS This article is based on a selective review of pertinent literature retrieved by a PubMed search. RESULTS Acute, severe headache, typically described as the worst headache of the patient's life, and meningismus are the characteristic manifestations of SAH. Computed tomog raphy (CT) reveals blood in the basal cisterns in the first 12 hours after SAH with approximately 95% sensitivity and specificity. If no blood is seen on CT, a lumbar puncture must be performed to confirm or rule out the diagnosis of SAH. All patients need intensive care so that rebleeding can be avoided and the sequelae of the initial bleed can be minimized. The immediate transfer of patients with acute SAH to a specialized center is crucially important for their outcome. In such centers, cerebral aneurysms can be excluded from the circulation either with an interventional endovascular procedure (coiling) or by microneurosurgery (clipping). CONCLUSION SAH is a life-threatening condition that requires immediate diagnosis, transfer to a neurovascular center, and treatment without delay.
Collapse
Affiliation(s)
- Athanasios K Petridis
- Department of Neurosurgery, Düsseldorf University Hospital; Department of Diagnostic and Interventional Radiology, Düsseldorf University Hospital; Department of Diagnostic and Interventional Radiology, Düsseldorf University Hospital
| | | | | | | | | | | | | |
Collapse
|
59
|
Ravid O, Elhaik Goldman S, Macheto D, Bresler Y, De Oliveira RI, Liraz-Zaltsman S, Gosselet F, Dehouck L, Beeri MS, Cooper I. Blood-Brain Barrier Cellular Responses Toward Organophosphates: Natural Compensatory Processes and Exogenous Interventions to Rescue Barrier Properties. Front Cell Neurosci 2018; 12:359. [PMID: 30459557 PMCID: PMC6232705 DOI: 10.3389/fncel.2018.00359] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 09/24/2018] [Indexed: 11/13/2022] Open
Abstract
Organophosphorus compounds (OPs) are highly toxic chemicals widely used as pesticides (e.g., paraoxon (PX)- the active metabolite of the insecticide parathion) and as chemical warfare nerve agents. Blood-brain barrier (BBB) leakage has been shown in rodents exposed to PX, which is an organophosphate oxon. In this study, we investigated the cellular mechanisms involved in BBB reaction after acute exposure to PX in an established in vitro BBB system made of stem-cell derived, human brain-like endothelial cells (BLECs) together with brain pericytes that closely mimic the in vivo BBB. Our results show that PX directly affects the BBB in vitro both at toxic and non-toxic concentrations by attenuating tight junctional (TJ) protein expression and that only above a certain threshold the paracellular barrier integrity is compromised. Below this threshold, BLECs exhibit a morphological coping mechanism in which they enlarge their cell area thus preventing the formation of meaningful intercellular gaps and maintaining barrier integrity. Importantly, we demonstrate that reversal of the apoptotic cell death induced by PX, by a pan-caspase-inhibitor ZVAD-FMK (ZVAD) can reduce PX-induced cell death and elevate cell area but do not prevent the induced BBB permeability, implying that TJ complex functionality is hindered. This is corroborated by formation of ROS at all toxic concentrations of PX and which are even higher with ZVAD. We suggest that while lower levels of ROS can induce compensating mechanisms, higher PX-induced oxidative stress levels interfere with barrier integrity.
Collapse
Affiliation(s)
- Orly Ravid
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Shirin Elhaik Goldman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - David Macheto
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Yael Bresler
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | | | - Sigal Liraz-Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory (LBHE), Université d'Artois, Lens, France
| | - Lucie Dehouck
- Blood-Brain Barrier Laboratory (LBHE), Université d'Artois, Lens, France
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Interdisciplinary Center Herzliya, Herzliya, Israel.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Interdisciplinary Center Herzliya, Herzliya, Israel
| |
Collapse
|
60
|
Li T, Xu W, Gao L, Guan G, Zhang Z, He P, Xu H, Fan L, Yan F, Chen G. Mesencephalic astrocyte-derived neurotrophic factor affords neuroprotection to early brain injury induced by subarachnoid hemorrhage via activating Akt-dependent prosurvival pathway and defending blood-brain barrier integrity. FASEB J 2018; 33:1727-1741. [PMID: 30211660 DOI: 10.1096/fj.201800227rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
This study aimed to explore the neuroprotective effect of mesencephalic astrocyte-derived neurotrophic factor (MANF) protein on early brain injury caused by subarachnoid hemorrhage (SAH) and the relevant mechanisms in experimental rats, expecting to understand whether MANF was a potential therapeutic target for SAH treatment. A perforation model of SAH was introduced into the study. Recombinant human MANF (rh-MANF) and protein kinase B (Akt) inhibitor (MK2206) were used to explore the effect and the mechanisms. Multiple approaches for systemic assessment were employed in the research, including the Garcia test, the SAH grade, Evans blue (EB) dye leakage, brain-water content (BWC), the rotarod test, and the Morris water-navigation task, as were biotechniques, such as immunohistochemistry, Western blot, transmission electron microscopy, and flow cytometry. MANF was mainly expressed in rat neurons, and its expression increased significantly at 3 h after SAH induction and peaked at 24 h. Stereotactic injection of rh-MANF into the cerebroventricle significantly increased the level of MANF, p-Akt, p-mouse double minute 2 homolog (p-MDM2), and B-cell lymphoma 2 (Bcl-2) in brain tissue, whereas it down-regulated the expression of P53, Bcl-2-associated X protein (Bax), and cleaved caspase-3, which indicated that neuronal apoptosis was remarkably suppressed. Expression of matrix metallopeptidase 9 (MMP-9) was also suppressed by the rh-MANF injection. Furthermore, neurologic deficits, EB dye leakage, and BWC were reduced, and long-lasting neuroprotection was noted with rh-MANF administration. The antiapoptotic and blood-brain barrier (BBB) protective effect could be offset by administering MK2206. MANF could alleviate neuronal apoptosis by activating Akt-dependent prosurvival pathway and abate BBB damage via MMP-9 suppression. MANF showed not only transient but also long-lasting neuroprotective properties. The rh-MANF as a potential drug for treating SAH might be of clinical use.-Li, T., Xu, W., Gao, L., Guan, G., Zhang, Z., He, P., Xu, H., Fan, L., Yan, F., Chen, G. Mesencephalic astrocyte-derived neurotrophic factor affords neuroprotection to early brain injury induced by subarachnoid hemorrhage via activating Akt-dependent prosurvival pathway and defending blood-brain barrier integrity.
Collapse
Affiliation(s)
- Tao Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guoping Guan
- Department of Neurosurgery, Fenghua Hospital, Ningbo, China
| | - Zhongyuan Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pingyou He
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hangzhe Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Linfeng Fan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
61
|
Schiefecker AJ, Rass V, Gaasch M, Kofler M, Thomé C, Humpel C, Ianosi B, Hackl WO, Beer R, Pfausler B, Schmutzhard E, Helbok R. Brain Extracellular Interleukin-6 Levels Decrease Following Antipyretic Therapy with Diclofenac in Patients with Spontaneous Subarachnoid Hemorrhage. Ther Hypothermia Temp Manag 2018; 9:48-55. [PMID: 30074854 DOI: 10.1089/ther.2018.0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In patients with aneurysmal subarachnoid hemorrhage (aSAH), increased brain extracellular interleukin (IL)-6 levels measured by cerebral microdialysis (CMD) were associated with disease severity, early brain injury, delayed cerebral infarction, and axonal injury. In this study, we analyzed brain extracellular IL-6 levels of aSAH patients following parenteral diclofenac. Twenty-four mechanically ventilated poor-grade aSAH patients were included. Changes in cerebral metabolism, brain/body temperature, and CMD-IL-6 levels following intravenous diclofenac infusion (DCF; 75 mg diluted in 100 cc normal saline) were retrospectively analyzed from prospectively collected bedside data (at 1 hour before DCF = baseline; and at 2, 4, and 8 hours after DCF). Statistical analysis was performed using generalized estimating equations. Seventy-two events in 24 aSAH patients were analyzed. Median age was 60 years (interquartile range [IQR]: 52-67), admission Hunt & Hess grade was 4 (IQR: 3-5), and modified Fisher grade (mFisher) was 4 (IQR: 3-4). Higher CMD-IL-6 levels at baseline were linked to fever, higher mFisher, delayed cerebral infarction, and metabolic distress (p < 0.05). CMD-IL-6 levels at baseline were 281.4 pg/mL (IQR: 47-1866) and significantly (p < 0.001; Wald-X2 = 106) decreased at 2 hours to 86.3 pg/mL (IQR: 7-1946), at 4 hours to 40.9 pg/mL (IQR: 4-1237), and at 8 hours to 53.5 pg/mL (IQR: 5-1085), independent of probe location or day after bleeding. Parenteral diclofenac may attenuate brain extracellular proinflammatory response in poor-grade aSAH patients.
Collapse
Affiliation(s)
- Alois J Schiefecker
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Rass
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Maxime Gaasch
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Mario Kofler
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudius Thomé
- 2 Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Humpel
- 3 Laboratory for Experimental Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria
| | - Bogdan Ianosi
- 4 Department of Medical Informatics and Technology, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall, Austria
| | - Werner O Hackl
- 4 Department of Medical Informatics and Technology, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall, Austria
| | - Ronny Beer
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Bettina Pfausler
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Erich Schmutzhard
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Raimund Helbok
- 1 Neurological Intensive Care Unit, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
62
|
Ma K, Li R, Zhao H, Qu J, Mu N, Liu X, Wang S, Yang C, Feng H, Tan L, Li F. Cattle Encephalon Glycoside and Ignotin Reduce Early Brain Injury and Cognitive Dysfunction after Subarachnoid Hemorrhage in Rats. Neuroscience 2018; 388:181-190. [PMID: 30036663 DOI: 10.1016/j.neuroscience.2018.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/01/2023]
Abstract
Subarachnoid hemorrhage (SAH) is a well-known hemorrhagic stroke with high rates of morbidity and mortality where patients frequently experience cognitive dysfunction. This study explores a potential treatment for cognitive dysfunction following SAH with the demonstration that multi-target drug cattle encephalon glycoside and ignotin (CEGI) can relieve cognitive dysfunction by decreasing hippocampal neuron apoptosis following SAH in rats. Experimentally, 110 male SD rats were separated at random into Sham (20), SAH + Vehicle (30), SAH + 4 ml/kg CEGI (30), and SAH + 1 ml/kg CEGI groups (30) and an endovascular perforation model was created to induce SAH. We discovered that the number of TUNEL-positive neurons in the hippocampus was markedly decreased in SAH + 4 ml/kg and SAH + 1 ml/kg CEGI groups compared to the SAH + Vehicle group. This finding was associated with an observed decrease in Bax/Bcl-2 ratio, cytochrome-c and PUMA expression, and the suppression of caspase-3 activation following SAH. In Morris water maze tests, the SAH + 4 ml/kg CEGI group demonstrated a decreased escape latency time and increase in time spent in the target quadrant as well as crossing times of platform region. These results indicate that high doses of CEGI can decrease hippocampal neuron apoptosis and relieve cognitive dysfunction in rats, suggesting that multitarget-drug CEGI exhibits a neuroprotective effect in SAH via the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Kang Ma
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Rongwei Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Qu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ning Mu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuanyan Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Fei Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
63
|
MST1 Suppression Reduces Early Brain Injury by Inhibiting the NF- κB/MMP-9 Pathway after Subarachnoid Hemorrhage in Mice. Behav Neurol 2018; 2018:6470957. [PMID: 30018671 PMCID: PMC6029491 DOI: 10.1155/2018/6470957] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/08/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
Background Mammalian sterile 20-like kinase 1 (MST1), the key component of the Hippo-YAP pathway, exhibits an important role in the pathophysiological process of various neurological disorders, including ischemic stroke and spinal cord injury. However, during subarachnoid hemorrhage, the involvement of MST1 in the pathophysiology of early brain injury remains unknown. Methods We employed intravascular filament perforation to establish the subarachnoid hemorrhage (SAH) mouse model. The MST1 inhibitor XMU-MP-1 was intraperitoneally injected at 1 h after SAH, followed by daily injections. MST1 in vivo knockdown was performed 3 weeks prior to SAH via intracerebroventricular injection of adeno-associated virus (AAV) packaged with MST1 shRNA. The SAH grade, behavioral deficits, TUNEL staining, Evans blue dye extravasation and fluorescence, brain water content, protein and cytokine expressions by Western blotting, immunofluorescence, and proteome cytokine array were evaluated. Results Following SAH, the phosphorylation level of MST1 was upregulated at 12 h, with a peak at 72 h after SAH. It was colocalized with the microglial marker Iba1. Both XMU-MP-1 and MST1 shRNA alleviated the neurological deficits, blood-brain barrier (BBB) disruption, brain edema, neuroinflammation, and white matter injury, which were induced by SAH in association with nuclear factor- (NF-) κB p65 and matrix metallopeptidase-9 (MMP-9) activation and downregulated endothelial junction protein expression. Conclusions The current findings indicate that MST1 participates in SAH-induced BBB disruption and white matter fiber damage via the downstream NF-κB-MMP-9 signaling pathway. Therefore, MST1 antagonists may serve as a novel therapeutic target to prevent early brain injury in SAH patients.
Collapse
|
64
|
Ma L, Jiang Y, Dong Y, Gao J, Du B, Liu D. Anti-TNF-alpha antibody attenuates subarachnoid hemorrhage-induced apoptosis in the hypothalamus by inhibiting the activation of Erk. Neuropsychiatr Dis Treat 2018; 14:525-536. [PMID: 29497296 PMCID: PMC5819588 DOI: 10.2147/ndt.s154809] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) can induce apoptosis in many regions of the brain including the cortex and hippocampus. However, few studies have focused on apoptosis in the hypothalamus after SAH. Although some antiapoptotic strategies have been developed for SAH, such as anti-tumor necrosis factor-alpha (TNF-α) antibody, the molecular mechanisms underlying this condition have yet to be elucidated. Therefore, the purpose of this study was to evaluate whether SAH could induce apoptosis in the hypothalamus and identify the potential molecular mechanisms underlying the actions of anti-TNF-α antibody, as a therapeutic regimen, upon apoptosis. MATERIALS AND METHODS SAH was induced in a rat model. Thirty minutes prior to SAH, anti-TNF-α antibody or U0126, an extracellular signal-regulated kinase (Erk) inhibitor, was microinjected into the left lateral cerebral ventricle. In addition, phorbol-12-myristate-13-acetate was injected intraperitoneally immediately after the anti-TNF-α antibody microinjection. Then, real-time polymerase chain reaction, Western blotting and immunohistochemistry were used to detect the expression of caspase-3, bax, bcl-2, phosphorylated Erk (p-Erk) and Erk. Finally, anxiety-like behavior was identified by using open field. RESULTS Levels of caspase-3, bax and bcl-2, all showed a temporary rise after SAH in the hypothalamus, indicating the induction of apoptosis in this brain region. Interestingly, we found that the microinjection of anti-TNF-α antibody could selectively block the elevated levels of bax, suggesting the potential role of anti-TNF-α antibody in the inhibition of SAH-induced apoptosis in the hypothalamus. Moreover, we found that Erk activation was necessary for apoptosis after SAH and that the microinfusion of anti-TNF-α antibody could inhibit apoptosis by suppressing the increase of p-Erk in the hypothalamus. Finally, our data indicated that the infusion of anti-TNF-α antibody could improve anxiety-like behavior. CONCLUSION Collectively, our data demonstrate that anti-TNF-α antibody attenuates apoptosis in the hypothalamus by inhibiting the activation of Erk, which plays an important role in the treatment of SAH.
Collapse
Affiliation(s)
- Ling Ma
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yong Jiang
- Department of Neurosurgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Yanan Dong
- Department of Neurosurgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Jun Gao
- Department of Neurosurgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Bin Du
- Department of Neurosurgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Dianwei Liu
- Department of Neurosurgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
65
|
Gu H, Wang YQ, Zhao CH, Zhong XM, Yang JG. The decrease of Tie-2 receptor phosphorylation in microvascular endothelial cells is involved in early brain injury after subarachnoid hemorrhage. Artery Res 2018. [DOI: 10.1016/j.artres.2018.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
66
|
Duris K, Splichal Z, Jurajda M. The Role of Inflammatory Response in Stroke Associated Programmed Cell Death. Curr Neuropharmacol 2018; 16:1365-1374. [PMID: 29473512 PMCID: PMC6251044 DOI: 10.2174/1570159x16666180222155833] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/17/2017] [Accepted: 02/22/2018] [Indexed: 01/13/2023] Open
Abstract
Stroke represents devastating pathology which is associated with a high morbidity and mortality. Initial damage caused directly by the onset of stroke, primary injury, may be eclipsed by secondary injury which may have a much more devastating effect on the brain. Primary injury is predominantly associated with necrotic cell death due to fatal insufficiency of oxygen and glucose. Secondary injury may on the contrary, lead apoptotic cell death due to structural damage which is not compatible with cellular functions or which may even represent the danger of malign transformation. The immune system is responsible for surveillance, defense and healing processes and the immune system plays a major role in triggering programmed cell death. Severe pathologies, such as stroke, are often associated with deregulation of the immune system, resulting in aggravation of secondary brain injury. The goal of this article is to overview the current knowledge about the role of immune system in the pathophysiology of stroke with respect to programmed neuronal cell death as well as to discuss current therapeutic strategies targeting inflammation after stroke.
Collapse
Affiliation(s)
| | | | - M. Jurajda
- Address correspondence to this author at the Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; E-mail:
| |
Collapse
|
67
|
Wang TH, Xiong LL, Yang SF, You C, Xia QJ, Xu Y, Zhang P, Wang SF, Liu J. LPS Pretreatment Provides Neuroprotective Roles in Rats with Subarachnoid Hemorrhage by Downregulating MMP9 and Caspase3 Associated with TLR4 Signaling Activation. Mol Neurobiol 2017; 54:7746-7760. [PMID: 27844284 DOI: 10.1007/s12035-016-0259-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023]
Abstract
Subarachnoid hemorrhage (SAH), as a severe brain disease, has high morbidity and mortality. SAH usually induced neurological dysfunction or death and the treatment is far from satisfaction. Here, we investigated the effect of low dose of LPS pretreatment and underlying molecular mechanism in rat SAH model. Firstly, SAH model was induced by prechiasmal cistern injection method (SAH1) and common carotid artery-prechiasmal cistern shunt method (SAH2), respectively, to select the more suitable SAH model. At 6, 12, 24, 48, and 72 h after SAH, brain injury including neurological dysfunction, blood-brain barrier disruption, brain edema, and cell apoptosis were detected. And the expression of MMP9, HMGB1/TLR4, and caspase3 in cortex were also explored. Then, SB-3CT, an inhibitor of MMP9, was administrated to investigate the exact function of MMP9 in the brain injury at 24 h after SAH. Moreover, low dose of LPS was used to verify whether it had nerve protection after SAH and the mechanism involving in MMP9 and caspase 3 was investigated. Our results showed SAH1 seems to be the most suitable SAH model. In addition, MMP9 activated by HMGB1/TLR4 may promote or aggravate brain injury, while inhibiting MMP9 via SB-3CT exerted a neuroprotective effect. Moreover, LPS improved the neurological dysfunction, reduced Evans blue extravasation and brain edema, and inhibited cell apoptosis of cortex in rats with brain injury induced by SAH. Importantly, LPS pretreatment increased the expression level of TLR4, and decreased the level of MMP9 and caspase3. Therefore, the present study revealed that low dose of LPS pretreatment could provide neuroprotective effects on brain injury caused by SAH via downregulating MMP9 and caspase3 and activating TLR4 signal pathway.
Collapse
Affiliation(s)
- Ting-Hua Wang
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Liu-Lin Xiong
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Shuai-Fen Yang
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Chao You
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Qing-Jie Xia
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yang Xu
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Piao Zhang
- Animal Zoology Department, Institute of Neuroscience, Kunming medical University, Kunming, 650000, China
| | - Shu-Fen Wang
- Yunnan Key Laboratory of Stem Cells and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, 650000, China.
| | - Jia Liu
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
- Animal Zoology Department, Institute of Neuroscience, Kunming medical University, Kunming, 650000, China.
| |
Collapse
|
68
|
Xu H, Li J, Wang Z, Feng M, Shen Y, Cao S, Li T, Peng Y, Fan L, Chen J, Gu C, Yan F, Wang L, Chen G. Methylene blue attenuates neuroinflammation after subarachnoid hemorrhage in rats through the Akt/GSK-3β/MEF2D signaling pathway. Brain Behav Immun 2017; 65:125-139. [PMID: 28457811 DOI: 10.1016/j.bbi.2017.04.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/14/2017] [Accepted: 04/25/2017] [Indexed: 12/12/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a serious medical problem with few effective pharmacotherapies available, and neuroinflammation has been identified as an important pathological process in early brain injury (EBI) after SAH. Methylene blue (MB) is an older drug that has been recently proven to exert extraordinary neuroprotective effects in several brain insults. However, no study has reported the beneficial effects of MB in SAH. In the current investigation, we studied the neuroprotective effects of MB in EBI after SAH and focused on its anti-inflammatory role. A total of 303 rats were subjected to an endovascular perforation process to produce an SAH model. We found that MB could significantly ameliorate brain edema secondary to BBB disruption and alleviate neurological dysfunction after SAH. MB administration also promoted the phosphorylation of Akt and GSK-3β, leading to an increased concentration of MEF2D in the nucleus. The cytokine IL-10 was up-regulated, and IL-1β, IL-6 and TNF-α were down-regulated after MB administration. MB administration could also alleviate neutrophil infiltration and microglia activation after SAH. MK2206, a selective inhibitor of Akt, abolished the neuroprotective effects of MB, inhibited the phosphorylation of Akt and prevented the nuclear localization of MEF2D. MK2206 also reduced the expression of IL-10 and increased the expression of pro-inflammatory cytokines. In conclusion, these data suggested that MB could ameliorate neuroinflammatory responses after SAH, and its anti-inflammatory effects might be exerted via activation of the Akt/GSK-3β/MEF2D pathway.
Collapse
Affiliation(s)
- Hangzhe Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Zhijiang Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Majing Feng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China; Department of Neurosurgery, Changxing People's Hospital, Taihuzhong Road 66th, Changxin, Huzhou 313100, China
| | - Yongfeng Shen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China; Department of Neurosurgery, Hangzhou First People's Hospital, Huansha Road 261st, Hangzhou 310006, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Tao Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Linfeng Fan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Jingyin Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Chi Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou 310016, China.
| |
Collapse
|
69
|
IMM-H004, A New Coumarin Derivative, Improved Focal Cerebral Ischemia via Blood–Brain Barrier Protection in Rats. J Stroke Cerebrovasc Dis 2017; 26:2065-2073. [DOI: 10.1016/j.jstrokecerebrovasdis.2016.11.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/21/2016] [Accepted: 11/25/2016] [Indexed: 01/04/2023] Open
|
70
|
Nrf2/HO-1 mediates the neuroprotective effect of mangiferin on early brain injury after subarachnoid hemorrhage by attenuating mitochondria-related apoptosis and neuroinflammation. Sci Rep 2017; 7:11883. [PMID: 28928429 PMCID: PMC5605716 DOI: 10.1038/s41598-017-12160-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023] Open
Abstract
Early brain injury (EBI) is involved in the process of cerebral tissue damage caused by subarachnoid hemorrhage (SAH), and multiple mechanisms, such as apoptosis and inflammation, participate in its development. Mangiferin (MF), a natural C-glucoside xanthone, has been reported to exert beneficial effects against several types of organ injury by influencing various biological progresses. The current study aimed to investigate the potential of MF to protect against EBI following SAH via histological and biological assessments. A rat perforation model of SAH was established, and MF was subsequently administered via intraperitoneal injection at a low and a high dose. High-dose MF significantly lowered the mortality of SAH animals and ameliorated their neurological deficits and brain edema. MF also dose-relatedly attenuated SAH-induced oxidative stress and decreased cortical cell apoptosis by influencing mitochondria-apoptotic proteins. In addition, MF downregulated the activation of the NLRP3 inflammasome and NF-κB as well as the production of inflammatory cytokines, and the expression of Nrf2 and HO-1 was upregulated by MF. The abovementioned findings indicate that MF is neuroprotective against EBI after SAH and Nrf2/HO-1 cascade may play a key role in mediating its effect through regulation of the mitochondrial apoptosis pathway and activation of the NLRP3 inflammasome and NF-κB.
Collapse
|
71
|
Yan F, Tan X, Wan W, Dixon BJ, Fan R, Enkhjargal B, Li Q, Zhang J, Chen G, Zhang JH. ErbB4 protects against neuronal apoptosis via activation of YAP/PIK3CB signaling pathway in a rat model of subarachnoid hemorrhage. Exp Neurol 2017; 297:92-100. [PMID: 28756200 DOI: 10.1016/j.expneurol.2017.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/17/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022]
Abstract
Neuronal apoptosis is a central pathological process in subarachnoid hemorrhage (SAH)-induced early brain injury. Previous studies indicated that ErbB4 (EGFR family member v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4) is essential for normal development and maintenance of the nervous system. In this study, we explored the neuroprotective effects of ErbB4 and its downstream YAP (yes-associated protein)/PIK3CB signaling pathway in early brain injury after SAH in a rat model using the endovascular perforation method. Rats were neurologically evaluated with the Modified Garcia Scale and beam balance test at 24h and 72h after SAH. An ErbB4 activator Neuregulin 1β1 (Nrg 1β1), ErbB4 siRNA and YAP siRNA were used to explore this pathway. The expression of p-ErbB4 and YAP was significantly increased after SAH. Multiple immunofluorescence labeling experiments demonstrated that ErbB4 is mainly expressed in neurons. Activation of ErbB4 and its downstream signals improved the neurological deficits after SAH and significantly reduced neuronal cell death. Inhibition of ErbB4 reduced YAP and PIK3CB expression, and aggravated cell apoptosis. YAP knockdown reduced the PIK3CB level and eliminated the anti-apoptotic effects of ErbB4 activation. These findings indicated that ErbB4 plays a neuroprotective role in early brain injury after SAH, possibly via the YAP/PIK3CB signaling pathway.
Collapse
Affiliation(s)
- Feng Yan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA; Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaoxiao Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Weifeng Wan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Ruiming Fan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Qian Li
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA.
| |
Collapse
|
72
|
Diniz MF, Ferreira DM, de Lima WG, Pedrosa ML, Silva ME, de Almeida Araujo S, Sampaio KH, de Campos TPR, Siqueira SL. Biodegradable seeds of holmium don't change neurological function after implant in brain of rats. Rep Pract Oncol Radiother 2017; 22:319-326. [PMID: 28663714 DOI: 10.1016/j.rpor.2017.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 02/12/2017] [Accepted: 03/27/2017] [Indexed: 01/24/2023] Open
Abstract
AIM To evaluate the surgical procedure and parenchymal abnormalities related to implantation of ceramic seeds with holmium-165 in rats' brain. BACKGROUND An effective method of cancer treatment is brachytherapy in which radioactive seeds are implanted in the tumor, generating a high local dose of ionizing radiation that can eliminate tumor cells while protecting the surrounding healthy tissue. Biodegradable Ho166-ceramic-seeds have been addressed recently. METHODS AND MATERIALS The experiments in this study were approved by the Ethics Committee on Animal Use at the Federal University of Ouro Preto, protocol number 2012/034. Twenty-one adult Fischer rats were divided into Naive Group, Sham Group and Group for seed implants (ISH). Surgical procedures for implantation of biodegradable seeds were done and 30 days after the implant radiographic examination and biopsy of the brain were performed. Neurological assays were also accomplished to exclude any injury resulting from either surgery or implantation of the seeds. RESULTS Radiographic examination confirmed the location of the seeds in the brain. Neurological assays showed animals with regular spontaneous activity. The histological analysis showed an increase of inflammatory cells in the brain of the ISH group. Electron microscopy evidenced cytoplasmic organelles to be unchanged. Biochemical analyzes indicate there was neither oxidative stress nor oxidative damage in the ISH brain. CAT activity showed no difference between the groups as well as lipid peroxidation measured by TBARS. CONCLUSIONS The analysis of the data pointed out that the performed procedure is safe as no animal showed alterations of the neurological parameters and the seeds did not promote histological architectural changes in the brain tissue.
Collapse
Affiliation(s)
- Mirla Fiuza Diniz
- Medical School, Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | | | | | - Maria Lucia Pedrosa
- Department of Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | | | | | | | | | | |
Collapse
|
73
|
Li T, Wang L, Hu Q, Liu S, Bai X, Xie Y, Zhang T, Bo S, Gao X, Wu S, Li G, Wang Z. Neuroprotective Roles of l-Cysteine in Attenuating Early Brain Injury and Improving Synaptic Density via the CBS/H 2S Pathway Following Subarachnoid Hemorrhage in Rats. Front Neurol 2017; 8:176. [PMID: 28512446 PMCID: PMC5411453 DOI: 10.3389/fneur.2017.00176] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/13/2017] [Indexed: 01/31/2023] Open
Abstract
l-Cysteine is a semi-essential amino acid and substrate for cystathionine-β-synthase (CBS) in the central nervous system. We previously reported that NaHS, an H2S donor, significantly alleviated brain damage after subarachnoid hemorrhage (SAH) in rats. However, the potential therapeutic value of l-cysteine and the molecular mechanism supporting these beneficial effects have not been determined. This study was designed to investigate whether l-cysteine could attenuate early brain injury following SAH and improve synaptic function by releasing endogenous H2S. Male Wistar rats were subjected to SAH induced by cisterna magna blood injection, and l-cysteine was intracerebroventricularly administered 30 min after SAH induction. Treatment with l-cysteine stimulated CBS activity in the prefrontal cortex (PFC) and H2S production. Moreover, l-cysteine treatment significantly ameliorated brain edema, improved neurobehavioral function, and attenuated neuronal cell death in the PFC; these effects were associated with a decrease in the Bax/Bcl-2 ratio and the suppression of caspase-3 activation 48 h after SAH. Furthermore, l-cysteine treatment activated the CREB-brain-derived neurotrophic factor (BDNF) pathway and intensified synaptic density by regulating synapse proteins 48 h after SAH. Importantly, all the beneficial effects of l-cysteine in SAH were abrogated by amino-oxyacetic acid, a CBS inhibitor. Based on these findings, l-cysteine may play a neuroprotective role in SAH by inhibiting cell apoptosis, upregulating CREB-BDNF expression, and promoting synaptic structure via the CBS/H2S pathway.
Collapse
Affiliation(s)
- Tong Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China.,Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Lingxiao Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China.,Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Quan Hu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China.,Department of Neurosurgery, Taian Central Hospital, Taian, China
| | - Song Liu
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Xuemei Bai
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Yunkai Xie
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Tiantian Zhang
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Shishi Bo
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Xiangqian Gao
- Department of Pathology, Binzhou Medical University Hospital, Binzhou, China
| | - Shuhua Wu
- Department of Pathology, Binzhou Medical University Hospital, Binzhou, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China
| | - Zhen Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
74
|
Thioredoxin-Interacting Protein Mediates Apoptosis in Early Brain Injury after Subarachnoid Haemorrhage. Int J Mol Sci 2017; 18:ijms18040854. [PMID: 28420192 PMCID: PMC5412438 DOI: 10.3390/ijms18040854] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/11/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023] Open
Abstract
Early brain injury (EBI) is considered to be the major factor associated with high morbidity and mortality after subarachnoid haemorrhage (SAH). Apoptosis is the major pathological mechanism of EBI, and its pathogenesis has not been fully clarified. Here, we report that thioredoxin-interacting protein (TXNIP), which is induced by protein kinase RNA-like endoplasmic reticulum (ER) kinase (PERK), participates in EBI by promoting apoptosis. By using adult male Sprague-Dawley rats to establish SAH models, as well as Terminal dexynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) staining, immunofluorescence, and western blot, we found that TXNIP expression significantly increased after SAH in comparison to the sham group and peaked at 48 h (up to 3.2-fold). Meanwhile, TXNIP was widely expressed in neurons and colocalized with TUNEL-positive cells in the hippocampus and cortex of SAH rats. After administration of TXNIP inhibitor-resveratrol (60 mg/kg), TXNIP small interfering RNA (siRNA) and the PERK inhibitor GSK2656157, TXNIP expression was significantly reduced, accompanied by an attenuation of apoptosis and prognostic indicators, including SAH grade, neurological deficits, brain water content, and blood-brain barrier (BBB) permeability. Collectively, these results suggest that TXNIP may participate in EBI after SAH by mediating apoptosis. The blockage of TXNIP induced by PERK could be a potential therapeutic strategy for SAH treatment.
Collapse
|
75
|
X-linked inhibitor of apoptosis inhibits apoptosis and preserves the blood-brain barrier after experimental subarachnoid hemorrhage. Sci Rep 2017; 7:44918. [PMID: 28327595 PMCID: PMC5361183 DOI: 10.1038/srep44918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 02/16/2017] [Indexed: 11/09/2022] Open
Abstract
Early brain injury following subarachnoid hemorrhage (SAH) strongly determines the prognosis of patients suffering from an aneurysm rupture, and apoptosis is associated with early brain injury after SAH. This study was designed to explore the role of X-linked inhibitor of apoptosis (XIAP) in early brain injury following SAH. The expression of XIAP was detected using western blotting and real-time RT-PCR in an autologous blood injection model of SAH. We also studied the role of XIAP in early brain injury and detected apoptosis-related proteins. The results showed that XIAP was significantly up-regulated in the cortex and hippocampus and that XIAP was mainly expressed in neuronal cells following SAH. The inhibition of endogenous XIAP aggravated blood-brain barrier disruption, neurological deficits and brain edema. Recombinant XIAP preserved the blood-brain barrier, improved the neurological scores and ameliorated brain edema. Recombinant XIAP treatment also decreased the expression of cleaved caspase-3, caspase-8 and caspase-9, whereas there was no effect on the expression of p53, apoptosis-inducing factor or cytochrome c. These results show that XIAP acts as an endogenous neuroprotective and anti-apoptotic agent following SAH. The effects of XIAP on early brain injury was associated with the inhibition of the caspase-dependent apoptosis pathway.
Collapse
|
76
|
Wang L, Li M, Xie Y, Xu L, Ye R, Liu X. Preclinical efficacy of human Albumin in subarachnoid hemorrhage. Neuroscience 2017; 344:255-264. [DOI: 10.1016/j.neuroscience.2016.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 11/26/2022]
|
77
|
van Lieshout JH, Dibué-Adjei M, Cornelius JF, Slotty PJ, Schneider T, Restin T, Boogaarts HD, Steiger HJ, Petridis AK, Kamp MA. An introduction to the pathophysiology of aneurysmal subarachnoid hemorrhage. Neurosurg Rev 2017; 41:917-930. [PMID: 28215029 DOI: 10.1007/s10143-017-0827-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 02/06/2023]
Abstract
Pathophysiological processes following subarachnoid hemorrhage (SAH) present survivors of the initial bleeding with a high risk of morbidity and mortality during the course of the disease. As angiographic vasospasm is strongly associated with delayed cerebral ischemia (DCI) and clinical outcome, clinical trials in the last few decades focused on prevention of these angiographic spasms. Despite all efforts, no new pharmacological agents have shown to improve patient outcome. As such, it has become clear that our understanding of the pathophysiology of SAH is incomplete and we need to reevaluate our concepts on the complex pathophysiological process following SAH. Angiographic vasospasm is probably important. However, a unifying theory for the pathophysiological changes following SAH has yet not been described. Some of these changes may be causally connected or present themselves as an epiphenomenon of an associated process. A causal connection between DCI and early brain injury (EBI) would mean that future therapies should address EBI more specifically. If the mechanisms following SAH display no causal pathophysiological connection but are rather evoked by the subarachnoid blood and its degradation production, multiple treatment strategies addressing the different pathophysiological mechanisms are required. The discrepancy between experimental and clinical SAH could be one reason for unsuccessful translational results.
Collapse
Affiliation(s)
- Jasper H van Lieshout
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| | - Maxine Dibué-Adjei
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Jan F Cornelius
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Philipp J Slotty
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Toni Schneider
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931, Köln, Germany
| | - Tanja Restin
- Zurich Centre for Integrative Human Physiology, Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.,Institute of Anesthesiology, Medical Faculty, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Hieronymus D Boogaarts
- Department of Neurosurgery, Medical Faculty, Radboud University Nijmegen, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Hans-Jakob Steiger
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Athanasios K Petridis
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Marcel A Kamp
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| |
Collapse
|
78
|
Chen F, Su X, Lin Z, Lin Y, Yu L, Cai J, Kang D, Hu L. Necrostatin-1 attenuates early brain injury after subarachnoid hemorrhage in rats by inhibiting necroptosis. Neuropsychiatr Dis Treat 2017; 13:1771-1782. [PMID: 28744127 PMCID: PMC5511017 DOI: 10.2147/ndt.s140801] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Necroptosis is programmed cell death that has been recently proposed and reported to be involved in several neurologic diseases. However, the role of necroptosis in early brain injury after subarachnoid hemorrhage (SAH) is still unknown. The purpose of this study was to investigate whether necroptosis was involved in SAH-induced early brain injury, and to assess the possible neuroprotective effect of necrostatin-1 using an endovascular perforation rat model of SAH. Our results showed that the expression levels of necroptosis-related proteins including RIP1, RIP3 and MLKL in the basal cortex all increased at 3 hours after SAH (P<0.05) and peaked at 48 hours after SAH (P<0.05). However, they were greatly reduced after treatment with necrostatin-1 (P<0.05). Concurrently, neurologic outcomes were significantly improved after necrostatin-1 treatment (P<0.05). Furthermore, brain edema, blood-brain barrier disruption, necrotic cell death and neuroinflammation were also greatly inhibited after necrostatin-1 treatment. These results indicate that necroptosis is an important mechanism of cell death involved in the early brain injury after experimental SAH. Necrostatin-1 perhaps can serve as a promising neuroprotective agent for SAH treatment.
Collapse
Affiliation(s)
- Fuxiang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xingfen Su
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhangya Lin
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yuanxiang Lin
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lianghong Yu
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jiawei Cai
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Dezhi Kang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Liwen Hu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
79
|
Choi KH, Kim HS, Park MS, Lee EB, Lee JK, Kim JT, Kim JH, Lee MC, Lee HJ, Cho KH. Overexpression of caveolin-1 attenuates brain edema by inhibiting tight junction degradation. Oncotarget 2016; 7:67857-67867. [PMID: 27708218 PMCID: PMC5356525 DOI: 10.18632/oncotarget.12346] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/25/2016] [Indexed: 02/07/2023] Open
Abstract
Cerebral edema from the disruption of the blood-brain barrier (BBB) after cerebral ischemia is a major cause of morbidity and mortality as well as a common event in patients with stroke. Caveolins (Cavs) are thought to regulate BBB functions. Here, we report for the first time that Cav-1 overexpression (OE) decreased brain edema from BBB disruption following ischemic insult. Edema volumes and Cav-1 expression levels were measured following photothrombosis and middle cerebral artery occlusion (MCAO). Endothelial cells that were transduced with a Cav-1 lentiviral expression vector were transplanted into rats. BBB permeability was quantified with Evans blue extravasation. Edema volume was determined from measures of the extravasation area, brain water content, and average fluorescence intensity after Cy5.5 injections. Tight junction (TJ) protein expression was measured with immunoblotting. Cav-1 expression levels and vasogenic brain edema correlated strongly after ischemic insult. Cav-1 expression and BBB disruption peaked 3 d after the MCAO. In addition, intravenous administration of endothelial cells expressing Cav-1 effectively increased the Cav-1 levels 3 d after the MCAO ischemic insult. Importantly, Cav-1 OE ameliorated the vasogenic edema by inhibiting the degradation of TJ protein expression in the acute phase of ischemic stroke. These results suggested that Cav-1 OE protected the integrity of the BBB mainly by preventing the degradation of TJ proteins in rats. These findings need to be confirmed in a clinical setting in human subjects.
Collapse
Affiliation(s)
- Kang-Ho Choi
- Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Man-Seok Park
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Eun-Bin Lee
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Jung-Kil Lee
- Department of Neurosurgery, Chonnam National University Medical School, Gwangju, Korea
| | - Joon-Tae Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Ja-Hae Kim
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Min-Cheol Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju, Korea
| | - Hong-Joon Lee
- Medical Research Institute, Chungang University College of Medicine, Seoul, Korea
| | - Ki-Hyun Cho
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
80
|
Affiliation(s)
- Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China;Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China, Collaborative Innovation Center for Brain Science, Hangzhou, Zhejiang, China
| | | |
Collapse
|
81
|
Atangana E, Schneider UC, Blecharz K, Magrini S, Wagner J, Nieminen-Kelhä M, Kremenetskaia I, Heppner FL, Engelhardt B, Vajkoczy P. Intravascular Inflammation Triggers Intracerebral Activated Microglia and Contributes to Secondary Brain Injury After Experimental Subarachnoid Hemorrhage (eSAH). Transl Stroke Res 2016; 8:144-156. [DOI: 10.1007/s12975-016-0485-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 07/11/2016] [Accepted: 07/14/2016] [Indexed: 12/29/2022]
|
82
|
Zhong YW, Wu J, Hu HL, Li WX, Zhong Y. Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy. Exp Ther Med 2016; 12:1908-1914. [PMID: 27588109 DOI: 10.3892/etm.2016.3526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/22/2016] [Indexed: 12/25/2022] Open
Abstract
Clinical studies have indicated that early brain injury (EBI) following subarachnoid hemorrhage (SAH) is associated with fatal outcomes. Oxidative stress and brain edema are the characteristic pathological events in occurrence EBI following SAH. The present study aimed to examine the effect of 3,4-dihydroxyphenylethanol (DOPET) against SAH-induced EBI, and to demonstrate whether the effect is associated with its potent free radical scavenging property. SAH was induced in rats using an endovascular perforation technique, and 24 h later the rats displayed diminished neurological scores and brain edema. Furthermore, elevated malondialdehyde (an index of lipid peroxidation) and depleted levels of antioxidants were observed in the rat cerebral cortex tissue. Quantitative polymerase chain reaction analysis indicated the upregulated mRNA expression of the apoptotic markers caspase-3 and -9 in the cerebral cortex. Furthermore, the protein expression levels of the proinflammatory cytokines tumor necrosis factor-α, interleukin (IL)-1β and IL-6 were significantly upregulated in SAH-induced rats. By constrast, treatment with DOPET significantly attenuated EBI by reducing brain edema, elevation of antioxidant status, inhibition of apoptosis and inflammation. In this context, DOPET may be a potent agent in the treatment of EBI following SAH, as a result of its free radical scavenging capacity.
Collapse
Affiliation(s)
- Yu-Wen Zhong
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Juan Wu
- Department of Obstetrics and Gynecology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Hua-Long Hu
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Wei-Xin Li
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Yong Zhong
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| |
Collapse
|
83
|
Caspase inhibitor zVAD-fmk protects against acute pancreatitis-associated lung injury via inhibiting inflammation and apoptosis. Pancreatology 2016; 16:733-8. [PMID: 27324074 DOI: 10.1016/j.pan.2016.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 05/08/2016] [Accepted: 06/03/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND/OBJECTIVES Pulmonary apoptosis is an important pathogenic mechanism of acute lung injury induced by many factors. This study aims to investigate whether the caspase inhibitor zVAD-fmk has a protective effect against lung injury in the severe acute pancreatitis model (SAP) in rats. METHODS Seventy-two Sprague-Dawley rats were randomly divided into Sham, SAP, and SAP + zVAD-fmk groups. The SAP model was established by injection of 5% sodium taurocholate into the pancreatic duct. Animals were sacrificed at 3 h, 6 h, 12 h, and 24 h after operation and then HE staining analysis was performed to assess the lung injury. ELISA was used to detect the activity of myeloperoxidase (MPO) and the concentrations of tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β). Western blotting was used to detect the expression of cleaved caspase-3 in the lung tissues. RESULTS Rats in SAP group showed obvious lung injury through pathologic examination. Pretreatment with zVAD-fmk significantly inhibited a post-SAP increase in the activation of MPO, TNF-α, IL-1β, and caspase-3, and decreased lung injury induced by SAP as determined by the pathologic score. CONCLUSION Our results suggest that apoptosis plays an important role in acute pancreatitis-associated lung injury (APALI), and inhibition of caspase activity may represent a new therapeutic approach for the treatment of APALI.
Collapse
|
84
|
Cao S, Zhu P, Yu X, Chen J, Li J, Yan F, Wang L, Yu J, Chen G. Hydrogen sulfide attenuates brain edema in early brain injury after subarachnoid hemorrhage in rats: Possible involvement of MMP-9 induced blood-brain barrier disruption and AQP4 expression. Neurosci Lett 2016; 621:88-97. [PMID: 27080433 DOI: 10.1016/j.neulet.2016.04.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 10/22/2022]
Abstract
AIMS This study investigated the effect of H2S on brain edema formation and the possible underlying mechanisms in early brain injury (EBI) of SAH using the endovascular perforation model. METHODS 96 male rats were randomly divided into four groups: sham group, SAH+vehicle group, SAH+low-dosage NaHS group, and SAH+high-dosage NaHS group. Brain samples were used for brain water content and blood-brain barrier (BBB) leakage measurement, gelatin zymography, Western blot and immunohistochemistry. RESULTS H2S markedly attenuated brain edema formation and apoptotic cell death, improved neurological dysfunction in the acute stage of SAH. The possible mechanisms of H2S's effect on brain edema formation were through preventing BBB disruption and reducing APQ4 expression on astrocytes. In detail, H2S prevented BBB disruption by inhibiting MMP-9 induced tight junction proteins (TJPs) degradation. H2S down-regulated AQP4 expression on astrocytes by suppressing glial cell activation and pro-inflammatory cytokines secretion. CONCLUSION Taken together, this study showed that H2S attenuated brain edema formation partially by inhibiting the degradation of TJPs via reducing MMP-9 expression/activity and suppressing AQP4 expression via alleviating glia activation and pro-inflammatory cytokines secretion.
Collapse
Affiliation(s)
- Shenglong Cao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Ping Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaobo Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jingyin Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jun Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China.
| |
Collapse
|
85
|
Provencio JJ, Swank V, Lu H, Brunet S, Baltan S, Khapre RV, Seerapu H, Kokiko-Cochran ON, Lamb BT, Ransohoff RM. Neutrophil depletion after subarachnoid hemorrhage improves memory via NMDA receptors. Brain Behav Immun 2016; 54:233-242. [PMID: 26872422 PMCID: PMC4828315 DOI: 10.1016/j.bbi.2016.02.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/29/2016] [Accepted: 02/08/2016] [Indexed: 01/17/2023] Open
Abstract
Cognitive deficits after aneurysmal subarachnoid hemorrhage (SAH) are common and disabling. Patients who experience delayed deterioration associated with vasospasm are likely to have cognitive deficits, particularly problems with executive function, verbal and spatial memory. Here, we report neurophysiological and pathological mechanisms underlying behavioral deficits in a murine model of SAH. On tests of spatial memory, animals with SAH performed worse than sham animals in the first week and one month after SAH suggesting a prolonged injury. Between three and six days after experimental hemorrhage, mice demonstrated loss of late long-term potentiation (L-LTP) due to dysfunction of the NMDA receptor. Suppression of innate immune cell activation prevents delayed vasospasm after murine SAH. We therefore explored the role of neutrophil-mediated innate inflammation on memory deficits after SAH. Depletion of neutrophils three days after SAH mitigates tissue inflammation, reverses cerebral vasoconstriction in the middle cerebral artery, and rescues L-LTP dysfunction at day 6. Spatial memory deficits in both the short and long-term are improved and associated with a shift of NMDA receptor subunit composition toward a memory sparing phenotype. This work supports further investigating suppression of innate immunity after SAH as a target for preventative therapies in SAH.
Collapse
Affiliation(s)
- Jose Javier Provencio
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA; Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA; Department of Neurology and Neuroscience, Brain Immunology and Glia Center, University of Virginia, PO Box 800394, Charlottesville, VA 22908, USA.
| | - Valerie Swank
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Haiyan Lu
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Sylvain Brunet
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Selva Baltan
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Rohini V Khapre
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Himabindu Seerapu
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Olga N Kokiko-Cochran
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Bruce T Lamb
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Richard M Ransohoff
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| |
Collapse
|
86
|
Choi KH, Kim HS, Park MS, Kim JT, Kim JH, Cho KA, Lee MC, Lee HJ, Cho KH. Regulation of Caveolin-1 Expression Determines Early Brain Edema After Experimental Focal Cerebral Ischemia. Stroke 2016; 47:1336-43. [DOI: 10.1161/strokeaha.116.013205] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/03/2016] [Indexed: 01/20/2023]
Abstract
Background and Purpose—
Most patients with cerebral infarction die of brain edema because of the breakdown of the blood–brain barrier (BBB) in ischemic tissue. Caveolins (a group of proteins) are key modulators of vascular permeability; however, a direct role of caveolin-1 (Cav-1) in the regulation of BBB permeability during ischemic injury has yet to be identified.
Methods—
Cav-1 expression was measured by immunoblotting after photothrombotic ischemia. A direct functional role of Cav-1 in cerebral edema and BBB permeability during cerebral ischemia was investigated by genetic manipulation (gene disruption and re-expression) of Cav-1 protein expression in mice.
Results—
There was a significant correlation between the extent of BBB disruption and the Cav-1 expression. In Cav-1–deficient (Cav-1
−/−
) mice, the extent of BBB disruption after cerebral ischemia was increased compared with wild-type (Cav-1
+/+
) mice, whereas the increase in cerebral edema volume was ameliorated by lentiviral-mediated re-expression of Cav-1. Furthermore, Cav-1
−/−
mice had significantly higher degradation of tight junction proteins and proteolytic activity of matrix metalloproteinase than Cav-1
+/+
mice. Conversely, re-expression of Cav-1 in Cav-1
−/−
mice restored tight junction protein expression and reduced matrix metalloproteinase proteolytic activity.
Conclusions—
These results indicate that Cav-1 is a critical determinant of BBB permeability. Strategies for regulating Cav-1 represent a novel therapeutic approach to controlling BBB disruption and subsequent neurological deterioration during cerebral ischemia.
Collapse
Affiliation(s)
- Kang-Ho Choi
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Hyung-Seok Kim
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Man-Seok Park
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Joon-Tae Kim
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Ja-Hae Kim
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Kyung-Ah Cho
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Min-Cheol Lee
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Hong-Joon Lee
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| | - Ki-Hyun Cho
- From the Department of Neurology, Chonnam National University Hwasun Hospital, Hwasun, Korea (K.-H.C.); Departments of Neurology (K.-H.C., M.-S.P., J.-T.K., K.-H.C.), Forensic Medicine (H.-S.K.), Nuclear Medicine (J.-H.K.), Biochemistry and Molecular Biology (K.-A.C.), and Pathology, Chonnam National University Medical School, Gwangju, Korea (M.-C.L.); and Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (H.-J.L.)
| |
Collapse
|
87
|
Abdullah Z, Bayraktutan U. Suppression of PKC-α attenuates TNF-α-evoked cerebral barrier breakdown via regulations of MMP-2 and plasminogen-plasmin system. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1354-66. [PMID: 27094771 DOI: 10.1016/j.bbadis.2016.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/08/2016] [Accepted: 03/30/2016] [Indexed: 10/21/2022]
Abstract
Ischaemic stroke, accompanied by neuroinflammation, impairs blood-brain barrier integrity through a complex mechanism involving both protein kinase C (PKC) and urokinase. Using an in vitro model of human blood-brain barrier (BBB) composed of brain microvascular endothelial cells (HBMEC) and astrocytes, this study assessed the putative roles of these elements in BBB damage evoked by enhanced availability of pro-inflammatory cytokine, TNF-α. Treatment of HBMEC with TNF-α significantly increased the mRNA and protein expressions of all plasminogen-plasmin system (PPS) components, namely tissue plasminogen activator, urokinase, urokinase plasminogen activator receptor and plasminogen activator inhibitor-1 and also the activities of urokinase, total PKC and extracellular MMP-2. Inhibition of urokinase by amiloride abated the effects of TNF-α on BBB integrity and MMP-2 activity without affecting that of total PKC. Conversely, pharmacological inhibition of conventional PKC isoforms dramatically suppressed TNF-α-induced overactivation of urokinase. Knockdown of PKC-α gene via specific siRNA in HBMEC suppressed the stimulatory effects of TNF-α on protein expression of all PPS components, MMP-2 activity, DNA fragmentation rates and pro-apoptotic caspase-3/7 activities. Establishment of co-cultures with BMEC transfected with PKC-α siRNA attenuated the disruptive effects of TNF-α on BBB integrity and function. This was partly due to elevations observed in expression of a tight junction protein, claudin-5 and partly to prevention of stress fibre formation. In conclusion, specific inhibition of PKC-α in cerebral conditions associated with exaggerated release of pro-inflammatory cytokines, notably TNF-α may be of considerable therapeutic value and help maintain endothelial cell viability, appropriate cytoskeletal structure and basement membrane.
Collapse
Affiliation(s)
- Zuraidah Abdullah
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham NG5 1PB, UK.
| |
Collapse
|
88
|
Saito A, Mekawy MM, Sumiyoshi A, Riera JJ, Shimizu H, Kawashima R, Tominaga T. Noninvasive targeting delivery and in vivo magnetic resonance tracking method for live apoptotic cells in cerebral ischemia with functional Fe2O3 magnetic nanoparticles. J Nanobiotechnology 2016; 14:19. [PMID: 26969152 PMCID: PMC4788935 DOI: 10.1186/s12951-016-0173-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/26/2016] [Indexed: 11/14/2022] Open
Abstract
Background Apoptotic neuronal death is known as programmed cell death. Inhibition of this progression might contribute to a new treatment strategy. However, methods for in vivo detection of live apoptotic cells are in need to be developed and established. Context and purpose The purpose of this study is to develop a new method for in vivo brain imaging for live apoptotic lesions using magnetic resonance imaging (MRI). We focused on the specific accumulation of our recently developed functional magnetic nanoparticles (FMNPs) into apoptotic cells using a rat cerebral ischemia model. Sulphorhodamine B, fluorescent dye was linked to valylalanylaspartic acid fluoromethyl ketone as a pan-caspase inhibitor to form SR-FLIVO. SR-FLIVO was bound with FMNPs to develop SR-FLIVO-FMNP probe. Ischemic rat brains were scanned by 7T MRI before and after intravenous injection of SR-FLIVO-FMNP and the distribution was evaluated by subtraction images of T2* colored mapping. SR-FLIVO, intracellular FMNPs, and T2* reduction area were histologically analyzed. The distribution of SR-FLIVO-FMNP was evaluated by subtracting the T2* signal images and was significantly correlated with the histological findings by TUNEL staining. Results Our experimental results revealed several findings where our newly developed probe SR-FLIVO-FMNP was intravenously administered into ischemic rats and FLIVO expression was tracked and found in apoptotic cells in rat brains after cerebral ischemia. A remarkable T2* reduction within the ischemic lesion was recorded using MRI based SR-FLIVO-FMNP probe as a contrasting agent due to the specific probe accumulation in apoptotic cells whereas, no observation of T2* reduction within the non-ischemic lesion due to no probe accumulation in non-apoptotic cells. Histological analysis based on the correlation between FLIVO and TUNEL staining showed that almost all FLIVO-positive cells were positive for TUNEL staining. These findings suggest the possibility for establishment of in vivo targeting delivery methods to live apoptotic cells based on conjugation of magnetic and fluorescent dual functional probes. Conclusion A newly developed probe SR-FLIVO-FMNP might be considered as a useful probe for in vivo apoptotic detection, and FMNPs might be a strong platform for noninvasive imaging and targeting delivery. Electronic supplementary material The online version of this article (doi:10.1186/s12951-016-0173-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Atsushi Saito
- Department of Neurosurgery, Aomori Prefectural Central Hospital, 2-1-1 Higashitsukurimichi, Aomori, 030-8553, Japan. .,Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Moataz M Mekawy
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan. .,National Institute for Materials Science, 1-Chome-2-1 Sengen, Tsukuba, Ibaraki Prefecture, 305-0047, Japan.
| | - Akira Sumiyoshi
- Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Jorge J Riera
- Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hiroaki Shimizu
- Department of Neurosurgery, Graduate School of Medicine, Akita University, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Ryuta Kawashima
- Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
89
|
Cakir M, Calikoglu C, Yılmaz A, Akpinar E, Bayraktutan Z, Topcu A. Serum nesfatin-1 levels: a potential new biomarker in patients with subarachnoid hemorrhage*. Int J Neurosci 2016; 127:154-160. [DOI: 10.3109/00207454.2016.1153473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Murteza Cakir
- Faculty of Medicine, Department of Neurosurgery, Ataturk University, Erzurum, Turkey
| | - Cagatay Calikoglu
- Faculty of Medicine, Department of Neurosurgery, Ataturk University, Erzurum, Turkey
| | - Atilla Yılmaz
- Faculty of Medicine, Department of Neurosurgery, Mustafa Kemal University, Hatay, Turkey
| | - Erol Akpinar
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, Turkey
| | - Zafer Bayraktutan
- Department of Biochemistry, Erzurum Region Education and Research Hospital, Erzurum, Turkey
| | - Atilla Topcu
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, Turkey
| |
Collapse
|
90
|
Yan F, Cao S, Li J, Dixon B, Yu X, Chen J, Gu C, Lin W, Chen G. Pharmacological Inhibition of PERK Attenuates Early Brain Injury After Subarachnoid Hemorrhage in Rats Through the Activation of Akt. Mol Neurobiol 2016; 54:1808-1817. [DOI: 10.1007/s12035-016-9790-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/11/2016] [Indexed: 12/17/2022]
|
91
|
Ma C, Pan Y, Yang Z, Meng Z, Sun R, Wang T, Fei Y, Fan W. Pre-administration of BAX-inhibiting peptides decrease the loss of the nigral dopaminergic neurons in rats. Life Sci 2016; 144:113-20. [DOI: 10.1016/j.lfs.2015.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/11/2015] [Accepted: 11/19/2015] [Indexed: 01/07/2023]
|
92
|
Rakkar K, Bayraktutan U. Increases in intracellular calcium perturb blood–brain barrier via protein kinase C-alpha and apoptosis. Biochim Biophys Acta Mol Basis Dis 2016; 1862:56-71. [DOI: 10.1016/j.bbadis.2015.10.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/14/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022]
|
93
|
Shishido H, Zhang H, Okubo S, Hua Y, Keep RF, Xi G. The Effect of Gender on Acute Hydrocephalus after Experimental Subarachnoid Hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:335-9. [PMID: 26463971 DOI: 10.1007/978-3-319-18497-5_58] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute hydrocephalus is a common complication of subarachnoid hemorrhage (SAH). We investigated the effect of gender on acute hydrocephalus development in a rat SAH model. SAH was induced in adult male and female Sprague-Dawley rats using endovascular perforation. Sham rats underwent the same procedure without perforation. Magnetic resonance imaging (MRI) was performed 24 h after SAH to determine ventricular volume. Hydrocephalus was defined as a ventricular volume that was more than 3 standard deviations from the mean value in sham-operated animals. After MRI, animals were euthanized and the extent of SAH was assessed using a modified grading system. No sham animals died. Mortality rates after SAH induction in male and female animals were 27 and 22 %, respectively. SAH induced significant ventricular enlargement compared with sham-operated rats (p < 0.01). The T2* hypointensity volume in the ventricle (used to assess intraventricular blood) was correlated with ventricular volume after SAH (r = 0.33, p < 0.05). The incidence of acute hydrocephalus 24 h after SAH was greater in female (75 %) than in male animals (47 %, p < 0.05) and the relative changes in ventricular volume were significantly larger in female than in male rats (292 ± 150 % vs 216 ± 127 % of sham-operated animals, respectively, p < 0.05). The increased hydrocephalus occurred even though SAH severity grade and ventricular T2* hypointensity volumes were not significantly different between male and female animals. Our data demonstrate that gender influences acute hydrocephalus development in a rat SAH model. Future studies should determine the role of estrogen in SAH-induced hydrocephalus.
Collapse
Affiliation(s)
- Hajime Shishido
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.,Department of Neurosurgery, Kagawa University, Kagawa, Japan
| | - Haining Zhang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.,Department of Neurology, 1st Affiliated Hospital, Jilin University, Changchun, China
| | - Shuichi Okubo
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.,Department of Neurosurgery, Kagawa University, Kagawa, Japan
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
94
|
Backer-Grøndahl A, Lindal S, Lorentzen MA, Eldevik P, Vorren T, Kristiansen B, Vangberg T, Ytrebø LM. A new non-craniotomy model of subarachnoid hemorrhage in the pig: a pilot study. Lab Anim 2015; 50:379-89. [PMID: 26643281 DOI: 10.1177/0023677215619806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Subarachnoid hemorrhage (SAH) from rupture of an intracranial arterial aneurysm is a devastating disease affecting young people, with serious lifelong disability or death as a frequent outcome. Large animal models that exhibit all the cardinal clinical features of human SAH are highly warranted. In this pilot study we aimed to develop a non-craniotomy model of SAH in pigs suitable for acute intervention studies. Six Norwegian Landrace pigs received advanced invasive hemodynamic and intracranial pressure (ICP) monitoring. The subarachnoid space, confirmed by a clear cerebrospinal fluid (CSF) tap, was reached by advancing a needle below the ocular bulb through the superior orbital fissure and into the interpeduncular cistern. SAH was induced by injecting 15 mL of autologous arterial blood into the subarachnoid space. Macro- and microanatomical investigations of the pig brain showed a typical blood distribution consistent with human aneurysmal SAH (aSAH) autopsy data. Immediately after SAH induction ICP sharply increased with a concomitant reduction in cerebral perfusion pressure (CPP). ICP returned to near normal values after 30 min, but increased subsequently during the experimental period. Signs of brain edema were confirmed by light microscopy post-mortem. None of the animals died during the experimental period. This new transorbital injection model of SAH in the pig mimics human aSAH and may be suitable for acute intervention studies. However, the model is technically challenging and needs further validation.
Collapse
Affiliation(s)
- Anders Backer-Grøndahl
- Department of Anesthesiology, University Hospital of North Norway, Tromsø, Norway Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Sigurd Lindal
- Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway Department of Pathology, University Hospital of North Norway, Tromsø, Norway
| | | | - Petter Eldevik
- Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Torgrim Vorren
- Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Bente Kristiansen
- Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Torgil Vangberg
- Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Lars Marius Ytrebø
- Department of Anesthesiology, University Hospital of North Norway, Tromsø, Norway Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
95
|
Teng Z, Jiang L, Hu Q, He Y, Guo Z, Wu Y, Huang Z, Cao F, Cheng C, Sun X, Guo Z. Peroxisome Proliferator-Activated Receptor β/δ Alleviates Early Brain Injury After Subarachnoid Hemorrhage in Rats. Stroke 2015; 47:196-205. [PMID: 26628385 DOI: 10.1161/strokeaha.115.011701] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Early brain injury is proposed to be the primary cause of the poor outcome after subarachnoid hemorrhage (SAH), which is closely related to the neural apoptosis. To date, the relationship between peroxisome proliferator-activated receptor β/δ (PPARβ/δ) and nuclear factor-κB/matrix metalloproteinase-9 (NF-κB/MMP-9) pathway, both of which are closely related to apoptotic effects, has been poorly studied in SAH. The present study was undertaken to evaluate the effects of PPARβ/δ on early brain injury and NF-κB/MMP-9 pathway after SAH in rats. METHODS SAH model was established by injecting nonheparinized autologous arterial blood into the prechiasmatic cistern in male Sprague-Dawley rats. Adenoviruses or small interfering RNAs were injected into the right lateral cerebral ventricle to, respectively, up- or downregulate PPARβ/δ expression before SAH. All animals were assessed with a neurological score and then killed at 24 hours after SAH surgery. The indexes of brain water content, blood-brain barrier permeability, and apoptosis were used to detect brain injury. The expression of PPARβ/δ, NF-κB, and MMP-9 were measured by immunohistochemistry, gelatin zymography, and Western Blot methods, respectively. In addition, GW0742, a specific agonist of PPARβ/δ, was used to treat SAH in rats, the effects of which were evaluated by neurological scoring and Evans blue extravasation. RESULTS Overexpression of PPARβ/δ by adenoviruses treatment significantly ameliorated brain injury with improvement in neurological deficits, brain edema, blood-brain barrier impairment, and neural cell apoptosis at 24 hours after SAH in rats, whereas downregulation of PPARβ/δ by small interfering RNAs administration resulted in the reverse effects of the above. The expression levels of NF-κB and MMP-9 were markedly downregulated when PPARβ/δ increased after PPARβ/δ adenovirus transfection and upregulated when PPARβ/δ decreased by PPARβ/δ small interfering RNAs treatment. Moreover, GW0742 improved neurological deficits and reduced Evans blue extravasation at 24 hours after SAH. CONCLUSIONS PPARβ/δ's overexpression may attenuate early brain injury after rats' SAH administration, which reduces neural apoptosis possibly through blocking NF-κB/MMP-9 pathway.
Collapse
Affiliation(s)
- Zhipeng Teng
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Li Jiang
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Qin Hu
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Yue He
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Zhenni Guo
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Yue Wu
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Zhijian Huang
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Fang Cao
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Chongjie Cheng
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Xiaochuan Sun
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo).
| | - Zongduo Guo
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo).
| |
Collapse
|
96
|
Yuan ZG, Wang JL, Jin GL, Yu XB, Li JQ, Qiu TL, Dai RX. Serum caspase-cleaved cytokeratin-18 levels and outcomes after aneurysmal subarachnoid hemorrhage. J Neurol Sci 2015; 359:298-304. [PMID: 26671131 DOI: 10.1016/j.jns.2015.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/03/2015] [Accepted: 11/11/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Cell apoptosis is involved in acute brain injury after aneurysmal subarachnoid hemorrhage (aSAH). The protein cytokeratin-18 (CK-18) is cleaved by the action of caspases during apoptosis, and the resulting fragments are released into the blood as caspase-cleaved CK (CCCK)-18. Our study examined the relationship between circulating CCCK-18 levels and long-term clinical outcomes among aSAH patients. METHODS We recruited 128 aSAH patients and 128 controls (matched on age and sex). Serum was collected at admission to the emergency department. Unfavorable outcome was defined as the Glasgow Outcome Score scores of 1-3. After a 6-month follow-up period, outcomes were assessed using a logistic regression analyses. The prognostic predictive values were evaluated according to receiver operating curves analysis. RESULTS aSAH patients had higher plasma CCCK-18 levels compared to controls (235.1 ± 86.8 U/L vs. 25.6 ± 23.4 U/L, P<0.001). CCCK-18 was independently associated with World Federation of Neurological Surgeons (WFNS) scores (t=4.460, P<0.001) and modified Fisher scores (t=3.781, P<0.001). Furthermore, CCCK-18 levels were markedly higher among patients with an unfavorable outcome and among non-survivors. CCCK-18 was yet identified as an independent prognostic predictor for mortality (odds ratio, 5.769; 95% confidence interval, 1.196-27.832; P=0.029) and unfavorable outcome (odds ratio, 4.909; 95% confidence interval, 1.521-15.838; P=0.008), as well as had similar predictive values for them compared with WFNS scores and modified Fisher scores. CONCLUSIONS High circulating CCCK-18 levels were associated with injury severity and a poor clinical outcome after aSAH and CCCK-18 had the potential to be a good prognostic biomarker for aSAH.
Collapse
Affiliation(s)
- Zi-Gang Yuan
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Jian-Li Wang
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China.
| | - Guo-Liang Jin
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Xue-Bin Yu
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Jin-Quan Li
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Tian-Lun Qiu
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Rong-Xiao Dai
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| |
Collapse
|
97
|
Shi SS, Zhang HB, Wang CH, Yang WZ, Liang RS, Chen Y, Tu XK. Propofol Attenuates Early Brain Injury After Subarachnoid Hemorrhage in Rats. J Mol Neurosci 2015; 57:538-45. [PMID: 26342279 DOI: 10.1007/s12031-015-0634-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/28/2015] [Indexed: 02/01/2023]
Abstract
Our previous studies demonstrated that propofol protects rat brain against focal cerebral ischemia. However, whether propofol attenuates early brain injury after subarachnoid hemorrhage in rats remains unknown until now. The present study was performed to evaluate the effect of propofol on early brain injury after subarachnoid hemorrhage in rats and further explore the potential mechanisms. Sprague-Dawley rats underwent subarachnoid hemorrhage (SAH) by endovascular perforation then received treatment with propofol (10 or 50 mg/kg) or vehicle after 2 and 12 h of SAH. SAH grading, neurological scores, brain water content, Evans blue extravasation, the myeloperoxidase activity, and malondialdehyde (MDA) content were measured 24 h after SAH. Expression of nuclear factor erythroid-related factor 2 (Nrf2), nuclear factor-kappa B (NF-κB) p65, and aquaporin 4 (AQP4) expression in rat brain were detected by Western blot. Expression of cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9) were determined by reverse transcription-polymerase chain reaction (RT-PCR). Expressions of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were assessed by ELISA. Neurological scores, brain water content, Evans blue extravasation, the myeloperoxidase activity, and MDA content were significantly reduced by propofol. Furthermore, expression of Nrf2 in rat brain was upregulated by propofol, and expression of NF-κB p65, AQP4, COX-2, MMP-9, TNF-α, and IL-1β in rat brain were attenuated by propofol. Our results demonstrated that propofol improves neurological scores, reduces brain edema, blood-brain barrier (BBB) permeability, inflammatory reaction, and lipid peroxidation in rats of SAH. Propofol exerts neuroprotection against SAH-induced early brain injury, which might be associated with the inhibition of inflammation and lipid peroxidation.
Collapse
Affiliation(s)
- Song-sheng Shi
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Hua-bin Zhang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Chun-hua Wang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wei-zhong Yang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Ri-sheng Liang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Ye Chen
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Xian-kun Tu
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
98
|
Fang L, Li X, Zhong Y, Yu J, Yu L, Dai H, Yan M. Autophagy protects human brain microvascular endothelial cells against methylglyoxal-induced injuries, reproducible in a cerebral ischemic model in diabetic rats. J Neurochem 2015; 135:431-40. [PMID: 26251121 DOI: 10.1111/jnc.13277] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/19/2015] [Accepted: 07/27/2015] [Indexed: 12/19/2022]
Abstract
Cerebral microvascular endothelial cells (ECs) are crucial for brain vascular repair and maintenance, but their physiological function may be impaired during ischemic stroke and diabetes. Methylglyoxal (MGO), a reactive dicarbonyl produced during glucose metabolism, could exacerbate ischemia-induced EC injury and dysfunction. We investigated the protective effect of autophagy on cultured human brain microvascular endothelial cells (HBMEC) that underwent MGO treatment. A further study was conducted to explore the underlying mechanisms of the protective effect. Autophagic activity was assessed by evaluating protein levels, using western blot. 3-methyladenine (3-MA), bafilomycin A1, ammonium chloride (AC), Beclin 1 siRNA, and chloroquine (CQ) were used to cause autophagy inhibition. Alarmar blue assay and lactate dehydrogenase release assay were used to evaluate cell viability. Streptozotocin was administered to induce type I diabetes in rats and post-permanent middle cerebral artery occlusion was performed to elicit cerebral ischemia. Blood-brain barrier permeability was also assessed. Our study found that MGO reduced HBMEC cell viability in a concentration- and time-dependent manner, and triggered the responsive autophagy activation. Autophagy inhibitors bafilomycin A1, AC, 3-MA, and BECN1 siRNA exacerbated MGO-induced HBMEC injury. FAK phosphorylation inhibitor PF573228 inhibited MGO-triggered autophagy and enhanced lactate dehydrogenase release. Meanwhile, similar autophagy activation in brain vascular ECs was observed during permanent middle cerebral artery occlusion-induced cerebral ischemia in diabetic rats, while chloroquine-induced autophagy inhibition enhanced blood-brain barrier permeability. Taken together, our study indicates that autophagy triggered by MGO defends HBMEC against injuries.
Collapse
Affiliation(s)
- Lili Fang
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinbo Zhong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Yu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lina Yu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haibin Dai
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Yan
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
99
|
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
|
100
|
Schneider UC, Davids AM, Brandenburg S, Müller A, Elke A, Magrini S, Atangana E, Turkowski K, Finger T, Gutenberg A, Gehlhaar C, Brück W, Heppner FL, Vajkoczy P. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathol 2015; 130:215-31. [PMID: 25956409 DOI: 10.1007/s00401-015-1440-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/17/2015] [Accepted: 05/01/2015] [Indexed: 01/08/2023]
Abstract
Inflammatory changes have been postulated to contribute to secondary brain injury after aneurysmal subarachnoid hemorrhage (SAH). In human specimens after SAH as well as in experimental SAH using mice, we show an intracerebral accumulation of inflammatory cells between days 4 and 28 after the bleeding. Using bone marrow chimeric mice allowing tracing of all peripherally derived immune cells, we confirm a truly CNS-intrinsic, microglial origin of these immune cells, exhibiting an inflammatory state, and rule out invasion of myeloid cells from the periphery into the brain. Furthermore, we detect secondary neuro-axonal injury throughout the time course of SAH. Since neuronal cell death and microglia accumulation follow a similar time course, we addressed whether the occurrence of activated microglia and neuro-axonal injury upon SAH are causally linked by depleting microglia in vivo. Given that the amount of neuronal cell death was significantly reduced after microglia depletion, we conclude that microglia accumulation inflicts secondary brain injury after SAH.
Collapse
Affiliation(s)
- Ulf C Schneider
- Department of Neurosurgery, Charité, Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|