1
|
Fu X, Ye F, Wan Y, Xi G, Hua Y, Keep RF. The Role of Complement C1qa in Experimental Intracerebral Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01302-4. [PMID: 39370487 DOI: 10.1007/s12975-024-01302-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
Evidence indicates that the complement system is activated and plays a role in brain injury after intracerebral hemorrhage (ICH). Most studies have focused on the role of C3, C5 and the membrane attack complex. The purpose of this study was to investigate the potential impact of complement C1q, a key upstream component of the classical pathway, on ICH-induced brain injury. Wild-type (WT) and C1qa knock out (KO) mice were compared using an autologous blood injection ICH model. Magnetic resonance imaging (MRI) was performed on days 1, 3 and 7 and brains harvested on days 3 and 7 for immunohistochemistry to examine brain injury mechanisms. WT and C1qa KO mice also received an intracerebral injection of thrombin, a key factor in ICH-induced brain injury. Following MRI scans, brains were harvested for immunohistochemistry on day 1. In comparison to WT mice, C1qa KO mice had reduced hematoma erythrolysis and neutrophil infiltration after ICH. However, they also had delayed hematoma clearance, which was associated with reduced induction of phagocytic multinuclear giant cells, and increased perihematomal neuronal damage. After thrombin injection, C1qa KO mice had smaller lesion volumes, less neuronal loss, reduced neutrophil infiltration, and less BBB damage. C1qa knockout has beneficial and detrimental effects on ICH-induced brain injury mechanisms, but a consistent beneficial effect after thrombin injection. Strategies to balance the roles of C1q after ICH may represent a promising therapeutic direction.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Yingfeng Wan
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
Zhang T, Xia F, Wan Y, Xi G, Ya H, Keep RF. Complement Inhibition Reduces Early Erythrolysis, Attenuates Brain Injury, Hydrocephalus, and Iron Accumulation after Intraventricular Hemorrhage in Aged Rats. Transl Stroke Res 2024:10.1007/s12975-024-01273-6. [PMID: 38943026 DOI: 10.1007/s12975-024-01273-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/03/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024]
Abstract
Blood components released by erythrolysis play an important role in secondary brain injury and posthemorrhagic hydrocephalus (PHH) after intraventricular hemorrhage (IVH). The current study examined the impact of N-acetylheparin (NAH), a complement inhibitor, on early erythrolysis, PHH and iron accumulation in aged rats following IVH. This study, on 18-months-old male Fischer 344 rats, was in 3 parts. First, rats had an intracerebroventricular injection of autologous blood (IVH) mixed with NAH or saline, or saline alone. After MRI at four hours, Western blot and immunohistochemistry examined complement activation and electron microscopy choroid plexus and periventricular damage. Second, rats had an IVH with NAH or vehicle, or saline. Rats underwent serial MRI at 4 h and 1 day to assess ventricular volume and erythrolysis. Immunohistochemistry and H&E staining examined secondary brain injury. Third, rats had an IVH with NAH or vehicle. Serial MRIs on day 1 and 28 assessed ventricular volume and iron accumulation. H&E staining and immunofluorescence evaluated choroid plexus phagocytes. Complement activation was found 4 h after IVH, and co-injection of NAH inhibited that activation. NAH administration attenuated erythrolysis, reduced ventricular volume, alleviated periventricular and choroid plexus injury at 4 h and 1 day after IVH. NAH decreased iron accumulation, the number of choroid plexus phagocytes, and attenuated hydrocephalus at 28 days after IVH. Inhibiting complement can reduce early erythrolysis, attenuates hydrocephalus and iron accumulation after IVH in aged animals.
Collapse
Affiliation(s)
- Tianjie Zhang
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Xia
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yingfeng Wan
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Hua Ya
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
3
|
Mavridis T, Choratta T, Papadopoulou A, Sawafta A, Archontakis-Barakakis P, Laou E, Sakellakis M, Chalkias A. Protease-Activated Receptors (PARs): Biology and Therapeutic Potential in Perioperative Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01233-0. [PMID: 38326662 DOI: 10.1007/s12975-024-01233-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Perioperative stroke is a devastating complication that occurs during surgery or within 30 days following the surgical procedure. Its prevalence ranges from 0.08 to 10% although it is most likely an underestimation, as sedatives and narcotics can substantially mask symptomatology and clinical presentation. Understanding the underlying pathophysiology and identifying potential therapeutic targets are of paramount importance. Protease-activated receptors (PARs), a unique family of G-protein-coupled receptors, are widely expressed throughout the human body and play essential roles in various physiological and pathological processes. This review elucidates the biology and significance of PARs, outlining their diverse functions in health and disease, and their intricate involvement in cerebrovascular (patho)physiology and neuroprotection. PARs exhibit a dual role in cerebral ischemia, which underscores their potential as therapeutic targets to mitigate the devastating effects of stroke in surgical patients.
Collapse
Affiliation(s)
- Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital, Dublin, incorporating the National Children's Hospital (AMNCH), Dublin, D24 NR0A, Ireland
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528, Athens, Greece
| | - Theodora Choratta
- Department of General Surgery, Metaxa Hospital, 18537, Piraeus, Greece
| | - Androniki Papadopoulou
- Department of Anesthesiology, G. Gennimatas General Hospital, 54635, Thessaloniki, Greece
| | - Assaf Sawafta
- Department of Cardiology, University Hospital of Larisa, 41110, Larisa, Greece
| | | | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, 15773, Athens, Greece
| | - Minas Sakellakis
- Department of Medicine, Jacobi Medical Center-North Central Bronx Hospital, Bronx, NY, 10467, USA
| | - Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104-5158, USA.
- Outcomes Research Consortium, Cleveland, OH, 44195, USA.
| |
Collapse
|
4
|
Ha GH, Yeon JY, Kim KH, Lee DM, Chae HY, Nam H, Lee K, Kim DO, Kim CK, Joo KM. Thrombin Priming Promotes the Neuroprotective Effects of Human Wharton's Jelly-Derived Mesenchymal Stem Cells Via the HGF/AKT/STAT3 Signaling Pathway. Stem Cells Dev 2024; 33:89-103. [PMID: 38164089 DOI: 10.1089/scd.2023.0191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Mesenchymal stem cells (MSCs) directly differentiate into neurons and endothelial cells after transplantation, and their secretome has considerable potential for treating brain injuries. Previous studies have suggested that the effects of MSCs priming with exposure to hypoxia, cytokines, growth factors, or chemical agents could optimize the paracrine potency and therapeutic potential of MSCs. Studies have suggested that thrombin-primed Wharton's Jelly-derived mesenchymal stem cells (Th.WJ-MSCs) significantly enhance the neuroprotective beneficial effects of naive MSCs in brain injury such as hypoxic-ischemic brain injury (HIE) and intraventricular hemorrhage (IVH). This study aimed to characterize WJ-MSCs in terms of stem cell markers, differentiation, cell proliferation, and paracrine factors by comparing naive and Th.WJ-MSCs. We demonstrated that compared with naive MSCs, Th.MSCs significantly enhanced the neuroprotective effects in vitro. Moreover, we identified differentially expressed proteins in the conditioned media of naive and Th.WJ-MSCs by liquid chromatography-tandem mass spectrometry analysis. Secretome analysis of the conditioned medium of WJ-MSCs revealed that such neuroprotective effects were mediated by paracrine effects with secretomes of Th.WJ-MSCs, and hepatocyte growth factor was identified as a key paracrine mediator. These results can be applied further in the preclinical and clinical development of effective and safe cell therapeutics for brain injuries such as HIE and IVH.
Collapse
Affiliation(s)
- Geun-Hyoung Ha
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Je Young Yeon
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ki Hoon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Du Man Lee
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Hye Yun Chae
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Hyun Nam
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kyunghoon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Dong Oh Kim
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Liang H, Zhang X, Hou Y, Zheng K, Hao H, He B, Li H, Sun C, Yang T, Song H, Cai R, Wang Y, Jiang H, Qi L, Wang Y. Super-high procoagulant activity of gecko thrombin: A gift from sky dragon. CNS Neurosci Ther 2023; 29:3081-3093. [PMID: 37144588 PMCID: PMC10493662 DOI: 10.1111/cns.14250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023] Open
Abstract
AIMS Gecko, the "sky dragon" named by Traditional Chinese Medicine, undergoes rapid coagulation and scarless regeneration following tail amputation in the natural ecology, providing a perfect opportunity to develop the efficient and safe drug for blood clotting. Here, gecko thrombin (gthrombin) was recombinantly prepared and comparatively studied on its procoagulant activity. METHODS The 3D structure of gthrombin was constructed using the homology modeling method of I-TASSER. The active gthrombin was prepared by the expression of gecko prethrombin-2 in 293 T cells, followed by purification with Ni2+ -chelating column chromatography prior to activation by snake venom-derived Ecarin. The enzymatic activities of gthrombin were assayed by hydrolysis of synthetic substrate S-2238 and the fibrinogen clotting. The vulnerable nerve cells were used to evaluate the toxicity of gthrombin at molecular and cellular levels. RESULTS The active recombinant gthrombin showed super-high catalytic and fibrinogenolytic efficiency than those of human under different temperatures and pH conditions. In addition, gthrombin made nontoxic effects on the central nerve cells including neurons, contrary to those of mammalian counterparts, which contribute to neuronal damage, astrogliosis, and demyelination. CONCLUSIONS A super-high activity but safe procoagulant candidate drug was identified from reptiles, which provided a promising perspective for clinical application in rapid blood clotting.
Collapse
Affiliation(s)
- Hao Liang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Xingyuan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Yuxuan Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Kang Zheng
- Anti‐aging & Regenerative Medicine Research Institution, School of Life Sciences and MedicineShandong University of TechnologyZiboPR China
| | - Huifei Hao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Hui Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Chunshuai Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Ting Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Rixin Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Haiyan Jiang
- Department of Emergency MedicineAffiliated Hospital of Nantong UniversityNantongPR China
| | - Lei Qi
- Department of Emergency MedicineAffiliated Hospital of Nantong UniversityNantongPR China
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| |
Collapse
|
6
|
Cheng J, Fan YQ, Zhang WF, Zhang G, Zeng K, Ye Z, Zhao D, Wu LQ, Chen ZB. Overexpressing SIRT6 can Attenuate the Injury of Intracerebral Hemorrhage by Down-Regulating NF-kB. Neuromolecular Med 2023; 25:53-63. [PMID: 35767210 DOI: 10.1007/s12017-022-08715-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 05/02/2022] [Indexed: 11/29/2022]
Abstract
Sirtuin-6 (SIRT6), a member of the sirtuins family of NAD ( +) dependent deacetylases, has been shown to have beneficial effects in ischemic stroke. However, the role of SIRT6 in intracerebral haemorrhage (ICH) has not reported. We observed that SIRT6 expression was down-regulated in human ICH patients and down-regulated in ICH-induced rat cortical neurons. We subsequently found that SIRT6 overexpression reduced brain tissue damage and increased neuronal survival in the ICH model of rats and hemin-induced cortical neurons. Our further study found that overexpression of SIRT6 can reduce inflammatory response by down-regulating the expression of NF-kB and thus promote the recovery of neurological function in ICH animals. In conclusion, SIRT6 can inhibit the expression of NF-kB and plays a neuroprotective role in ICH by inhibiting the NF-kB-mediated inflammatory response.SIRT6 could be a novel therapeutic target for ICH.
Collapse
Affiliation(s)
- Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Yan-Qin Fan
- Department of Nephrology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Wen-Fei Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Guo Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Kuo Zeng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Zhang Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Dan Zhao
- The Open Project of Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Li-Quan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Zhi-Biao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China.
| |
Collapse
|
7
|
Zuo W, Wang Y, Sun J, Zhang Y. Effects and mechanism of myeloperoxidase on microglia in the early stage of intracerebral hemorrhage. Front Neurosci 2022; 16:1046244. [PMID: 36570834 PMCID: PMC9783921 DOI: 10.3389/fnins.2022.1046244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
Objectives (1) To clarify the dynamic relationship between the expression of myeloperoxidase (MPO) and microglial activation of intracerebral hemorrhage (ICH), (2) to explore the effect of inhibition of MPO on microglial activation, and (3) to observe the improvement in the neurobehavior of mice with inhibition of MPO. Methods C57 BL/6 mice and CX3CR1 + /GFP mice were used to establish a phosphate-buffered saline (PBS) group, an ICH group, and a 4-aminobenzoic acid hydrazide (ABAH) group. Longa score, open field locomotion, hind-limb clasping test, immunohistochemistry, immunofluorescence, blood routine detection, and flow cytometry were used. Results The neurobehavior of the mice was significantly impaired following ICH (P < 0.01); the expression of MPO was significantly increased following ICH, and reached a peak value at 6 h post-injury (P < 0.001). Moreover, the microglial activation increased significantly following ICH, and reached a peak level at 24 h post-injury (P < 0.01). Following inhibition of MPO, the activation of microglia in the ICH group decreased significantly (P < 0.001). Moreover, the neurobehavior of the ICH group was significantly improved with MPO inhibition (P < 0.05). Conclusion MPO may be an upstream molecule activated by microglia and following inhibition of MPO can improve secondary injury resulting from ICH.
Collapse
Affiliation(s)
- Wei Zuo
- Department of Neuro-Oncological Surgery, Neurosurgery Center, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yunchang Wang
- Xiangya Hospital, Central Southern University, Changsha, China,Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiali Sun
- College of Life Sciences, Central Southern University, Changsha, China
| | - Yinian Zhang
- Department of Neuro-Oncological Surgery, Neurosurgery Center, Zhujiang Hospital of Southern Medical University, Guangzhou, China,Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China,*Correspondence: Yinian Zhang,
| |
Collapse
|
8
|
Zhang Z, Tan Q, Guo P, Huang S, Jia Z, Liu X, Feng H, Chen Y. NLRP3 inflammasome-mediated choroid plexus hypersecretion contributes to hydrocephalus after intraventricular hemorrhage via phosphorylated NKCC1 channels. J Neuroinflammation 2022; 19:163. [PMID: 35729645 PMCID: PMC9210649 DOI: 10.1186/s12974-022-02530-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 06/14/2022] [Indexed: 12/26/2022] Open
Abstract
Background Hydrocephalus is a severe complication of intracerebral hemorrhage with ventricular extension (ICH-IVH) and causes cerebrospinal fluid (CSF) accumulation. The choroid plexus epithelium plays an important role in CSF secretion and constitutes the blood–CSF barrier within the brain–immune system interface. Although the NLRP3 inflammasome, as a key component of the innate immune system, promotes neuroinflammation, its role in the pathogenesis of hydrocephalus after hemorrhage has not been investigated. Therefore, this study aimed to investigate the potential mechanism of NLRP3 in hydrocephalus to discover a potential marker for targeted therapy. Methods A rat model of hydrocephalus after ICH-IVH was developed through autologous blood infusion in wild-type and Nlrp3−/− rats. By studying the features and processes of the model, we investigated the relationship between the NLRP3 inflammasome and CSF hypersecretion in the choroid plexus. Results The ICH-IVH model rats showed ventricular dilation accompanied by CSF hypersecretion for 3 days. Based on the choroid plexus RNA-seq and proteomics results, we found that an inflammatory response was activated. The NLRP3 inflammasome was investigated, and the expression levels of NLRP3 inflammasome components reached a peak at 3 days after ICH-IVH. Inhibition of NLRP3 by an MCC950 inflammasome inhibitor or Nlrp3 knockout decreased CSF secretion and ventricular dilation and attenuated neurological deficits after ICH-IVH. The mechanism underlying the neuroprotective effects of NLRP3 inhibition involved decreased phosphorylation of NKCC1, which is a major protein that regulates CSF secretion by altering Na+- and K+-coupled water transport, via MCC950 or Nlrp3 knockout. In combination with the in vitro experiments, this experiment confirmed the involvement of the NLRP3/p-NKCC1 pathway and Na+ and K+ flux. Conclusions This study demonstrates that NKCC1 phosphorylation in the choroid plexus epithelium promotes NLRP3 inflammasome-mediated CSF hypersecretion and that NLRP3 plays an important role in the pathogenesis of hydrocephalus after hemorrhage. These findings provide a new therapeutic strategy for treating hydrocephalus. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02530-x.
Collapse
Affiliation(s)
- Zhaoqi Zhang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qiang Tan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Suna Huang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhengcai Jia
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| |
Collapse
|
9
|
Zhou G, Fu X, Wang L, Cao Y, Zhuang J, Hu J, Li Y, Xu C, Gao S, Shao A, Wang L. Palmitoylethanolamide ameliorates neuroinflammation via modulating PPAR-α to promote the functional outcome after intracerebral hemorrhage. Neurosci Lett 2022; 781:136648. [DOI: 10.1016/j.neulet.2022.136648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 01/23/2023]
|
10
|
Physical Exercise as a Modulator of Vascular Pathology and Thrombin Generation to Improve Outcomes After Traumatic Brain Injury. Mol Neurobiol 2021; 59:1124-1138. [PMID: 34846694 DOI: 10.1007/s12035-021-02639-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/04/2021] [Indexed: 10/19/2022]
Abstract
Disruption of the blood-brain barrier and occurrence of coagulopathy after traumatic brain injury (TBI) have important implications for multiple secondary injury processes. Given the extent of post-traumatic changes in neuronal function, significant alterations in some targets, such thrombin (a protease that plays a physiological role in maintaining blood coagulation), play an important role in TBI-induced pathophysiology. Despite the magnitude of thrombin in synaptic plasticity being concentration-dependent, the mechanisms underlying TBI have not been fully elucidated. The understanding of this post-injury neurovascular dysregulation is essential to establish scientific-based rehabilitative strategies. One of these strategies may be supporting physical exercise, considering its relevance in reducing damage after a TBI. However, there are caveats to consider when interpreting the effect of physical exercise on neurovascular dysregulation after TBI. To complete this picture, this review will describe how the interactions established between blood-borne factors (such as thrombin) and physical exercise alter the TBI pathophysiology.
Collapse
|
11
|
Vittal Rao H, Bihaqi SW, Iannucci J, Sen A, Grammas P. Thrombin Signaling Contributes to High Glucose-Induced Injury of Human Brain Microvascular Endothelial Cells. J Alzheimers Dis 2021; 79:211-224. [PMID: 33252072 DOI: 10.3233/jad-200658] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Diabetes is one of the strongest disease-related risk factors for Alzheimer's disease (AD). In diabetics, hyperglycemia-induced microvascular complications are the major cause of end-organ injury, contributing to morbidity and mortality. Microvascular pathology is also an important and early feature of AD. The cerebral microvasculature may be a point of convergence of both diseases. Several lines of evidence also implicate thrombin in AD as well as in diabetes. OBJECTIVE Our objective was to investigate the role of thrombin in glucose-induced brain microvascular endothelial injury. METHODS Cultured Human brain microvascular endothelial cells (HBMVECs) were treated with 30 mM glucose±100 nM thrombin and±250 nM Dabigatran or inhibitors of PAR1, p38MAPK, MMP2, or MMP9. Cytotoxicity and thrombin activity assays on supernatants and western blotting for protein expression in lysates were performed. RESULTS reatment of HBMVECs with 30 mM glucose increased thrombin activity and expression of inflammatory proteins TNFα, IL-6, and MMPs 2 and 9; this elevation was reduced by the thrombin inhibitor dabigatran. Direct treatment of brain endothelial cells with thrombin upregulated p38MAPK and CREB, and induced TNFα, IL6, MMP2, and MMP9 as well as oxidative stress proteins NOX4 and iNOS. Inhibition of thrombin, thrombin receptor PAR1 or p38MAPK decrease expression of inflammatory and oxidative stress proteins, implying that thrombin may play a central role in glucose-induced endothelial injury. CONCLUSION Since preventing brain endothelial injury would preserve blood-brain barrier integrity, prevent neuroinflammation, and retain intact functioning of the neurovascular unit, inhibiting thrombin, or its downstream signaling effectors, could be a therapeutic strategy for mitigating diabetes-induced dementia.
Collapse
Affiliation(s)
- Haripriya Vittal Rao
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Syed Waseem Bihaqi
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA.,Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Jaclyn Iannucci
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Abhik Sen
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA.,Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Paula Grammas
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
12
|
Wang M, Xia F, Wan S, Hua Y, Keep RF, Xi G. Role of Complement Component 3 in Early Erythrolysis in the Hematoma After Experimental Intracerebral Hemorrhage. Stroke 2021; 52:2649-2660. [PMID: 34176310 DOI: 10.1161/strokeaha.121.034372] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ming Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.).,Brain Center, Zhejiang Hospital, Zhejiang University Medical School, Hangzhou, China (M.W., S.W.)
| | - Fan Xia
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| | - Shu Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.).,Brain Center, Zhejiang Hospital, Zhejiang University Medical School, Hangzhou, China (M.W., S.W.)
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| |
Collapse
|
13
|
Ye F, Hua Y, Keep RF, Xi G, Garton HJL. CD47 blocking antibody accelerates hematoma clearance and alleviates hydrocephalus after experimental intraventricular hemorrhage. Neurobiol Dis 2021; 155:105384. [PMID: 33945877 DOI: 10.1016/j.nbd.2021.105384] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/26/2021] [Accepted: 04/30/2021] [Indexed: 12/22/2022] Open
Abstract
Background CD47, a glycoprotein on red blood cell membranes, inhibits phagocytosis via interaction with signal regulatory protein α on phagocytes. Our previous research has demonstrated that blocking CD47 accelerates hematoma clearance and reduces brain injury after intracerebral hemorrhage. The current study investigated whether phagocytosis or erythrocyte CD47 impacts hematoma resolution and hydrocephalus development after intraventricular hemorrhage (IVH). Methods Adult (3-month-old) male Fischer 344 rats were intraventricularly injected with 200 μl autologous blood, mixed with either CD47 blocking antibody or isotype IgG, or 200 μl saline as control. In subgroups of CD47 blocking antibody treated rats, clodronate liposomes (to deplete microglia/monocyte-derived macrophages) or control liposomes were co-injected. Magnetic resonance imaging (MRI) was used to evaluate ventricular volume and intraventricular T2* lesion volume (estimating hematoma volume). The brains were harvested after 4 or 72 h for histology to evaluate phagocytosis. Results In adult male rats, CD47 blocking antibody alleviated hydrocephalus development by day 3. In addition, the CD47 blocking antibody reduced intraventricular T2* lesion and T2* non-hypointense lesion size after IVH through day 1 to day 3. Erythrophagocytosis was observed as soon as 4 h after IVH and was enhanced on day 3. Furthermore, intra-hematoma infiltration of CD68, heme oxygenase-1 and ferritin positive phagocytes were upregulated by CD47 blockade by day 3. Clodronate liposomes co-injection caused more severe hydrocephalus and weight loss. Conclusion Blocking CD47 in the hematoma accelerated hematoma clearance and alleviated hemolysis and hydrocephalus development after IVH, suggesting CD47 might be valuable in the future treatment for IVH.
Collapse
Affiliation(s)
- Fenghui Ye
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - 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.
| | - Hugh J L Garton
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Shlobin NA, Har-Even M, Itsekson-Hayosh Z, Harnof S, Pick CG. Role of Thrombin in Central Nervous System Injury and Disease. Biomolecules 2021; 11:562. [PMID: 33921354 PMCID: PMC8070021 DOI: 10.3390/biom11040562] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Thrombin is a Na+-activated allosteric serine protease of the chymotrypsin family involved in coagulation, inflammation, cell protection, and apoptosis. Increasingly, the role of thrombin in the brain has been explored. Low concentrations of thrombin are neuroprotective, while high concentrations exert pathological effects. However, greater attention regarding the involvement of thrombin in normal and pathological processes in the central nervous system is warranted. In this review, we explore the mechanisms of thrombin action, localization, and functions in the central nervous system and describe the involvement of thrombin in stroke and intracerebral hemorrhage, neurodegenerative diseases, epilepsy, traumatic brain injury, and primary central nervous system tumors. We aim to comprehensively characterize the role of thrombin in neurological disease and injury.
Collapse
Affiliation(s)
- Nathan A. Shlobin
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Meirav Har-Even
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ze’ev Itsekson-Hayosh
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel;
- Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer 5262000, Israel
| | - Sagi Harnof
- Department of Neurosurgery, Beilinson Hospital, Rabin Medical Center, Tel Aviv University, Petah Tikva 4941492, Israel;
| | - Chaim G. Pick
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Center for Biology of Addictive Diseases, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
15
|
Deng X, Wu S, Li Z, Zhao Y, Duan C. Ratiometric Detection of DNA and Protein in Serum by a Universal Tripyridinyl RuII Complex–Encapsulated SiO2@Polydopamine Fluorescence Nanoplatform. Anal Chem 2020; 92:15908-15915. [DOI: 10.1021/acs.analchem.0c03306] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Xunxun Deng
- School of Chemical Engineering, Dalian University of Technology, Dalian 116023, PR China
- Zhangdayu School of Chemistry, Dalian University of Technology, Dalian 116023, PR China
| | - Shuo Wu
- School of Chemical Engineering, Dalian University of Technology, Dalian 116023, PR China
| | - Zhipeng Li
- School of Chemical Engineering, Dalian University of Technology, Dalian 116023, PR China
| | - Yanqiu Zhao
- School of Chemical Engineering, Dalian University of Technology, Dalian 116023, PR China
| | - Chunying Duan
- Zhangdayu School of Chemistry, Dalian University of Technology, Dalian 116023, PR China
| |
Collapse
|
16
|
Jamei HR, Rezaei B, Ensafi AA. Ultra-sensitive and selective electrochemical biosensor with aptamer recognition surface based on polymer quantum dots and C 60/MWCNTs- polyethylenimine nanocomposites for analysis of thrombin protein. Bioelectrochemistry 2020; 138:107701. [PMID: 33254052 DOI: 10.1016/j.bioelechem.2020.107701] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
In this study, an ultra-sensitive and selective Thrombin biosensor with aptamer-recognition surface is introduced based on carbon nanocomposite. To prepare the this biosensor, screen-printed carbon electrodes (SPCE) were modified with a nanocomposite made from fullerene (C60), multi-walled carbon nanotubes (MWCNTs), polyethylenimine (PEI) and polymer quantum dots (PQdot). The unique characteristics of each component of the C60/MWCNTs-PEI/PQdot nanocomposite allow for synergy between nanoparticles while polymer quantum dots resulted in characteristics such as high stability, high surface to volume ratio, high electrical conductivity, high biocompatibility, and high mechanical and chemical stability. The large number of amine groups in C60/MWCNTs-PEI/PQdot nanocomposite created more sites for better covalent immobilization of amino-linked aptamer (APT) which improved the sensitivity and stability of the aptasensor. Differential Pulse Voltammetry (DPV) method with probe solution was used as the measurment method. Binding of thrombin protein to aptamers immobilized on the transducer resulted in reduced electron transfer at the electrode/electrolyte interface which reduces the peak current (IP) in DPV. The calibration curve was drawn using the changes in the peak current (ΔIP),. The proposed aptasensor has a very low detection limit of 6 fmol L-1, and a large linear range of 50 fmol L-1 to 20 nmol L-1. Furthermore, the proposed C60/MWCNTs-PEI/PQdot/APT aptasensor has good reproducibility, great selectivity, low response time and a good stability during its storage. Finally, the application of the proposed aptasensor for measuring thrombin on human blood serum samples was investigated. This aptasensor can be useful in bioengineering and biomedicine applications as well as for clinical studies.
Collapse
Affiliation(s)
- Hamid Reza Jamei
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Behzad Rezaei
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Ali Asghar Ensafi
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
17
|
Ye F, Garton HJL, Hua Y, Keep RF, Xi G. The Role of Thrombin in Brain Injury After Hemorrhagic and Ischemic Stroke. Transl Stroke Res 2020; 12:496-511. [PMID: 32989665 DOI: 10.1007/s12975-020-00855-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023]
Abstract
Thrombin is increased in the brain after hemorrhagic and ischemic stroke primarily due to the prothrombin entry from blood either with a hemorrhage or following blood-brain barrier disruption. Increasing evidence indicates that thrombin and its receptors (protease-activated receptors (PARs)) play a major role in brain pathology following ischemic and hemorrhagic stroke (including intracerebral, intraventricular, and subarachnoid hemorrhage). Thrombin and PARs affect brain injury via multiple mechanisms that can be detrimental or protective. The cleavage of prothrombin into thrombin is the key step of hemostasis and thrombosis which takes place in every stroke and subsequent brain injury. The extravascular effects and direct cellular interactions of thrombin are mediated by PARs (PAR-1, PAR-3, and PAR-4) and their downstream signaling in multiple brain cell types. Such effects include inducing blood-brain-barrier disruption, brain edema, neuroinflammation, and neuronal death, although low thrombin concentrations can promote cell survival. Also, thrombin directly links the coagulation system to the immune system by activating interleukin-1α. Such effects of thrombin can result in both short-term brain injury and long-term functional deficits, making extravascular thrombin an understudied therapeutic target for stroke. This review examines the role of thrombin and PARs in brain injury following hemorrhagic and ischemic stroke and the potential treatment strategies which are complicated by their role in both hemostasis and brain.
Collapse
Affiliation(s)
- Fenghui Ye
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Hugh J L Garton
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
18
|
Wogonin Accelerates Hematoma Clearance and Improves Neurological Outcome via the PPAR-γ Pathway After Intracerebral Hemorrhage. Transl Stroke Res 2020; 12:660-675. [PMID: 32918259 DOI: 10.1007/s12975-020-00842-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 07/22/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
Intracerebral hemorrhage (ICH) is a cerebrovascular disease with high mortality and morbidity for which effective treatments are currently lacking. Wogonin is a major flavonoid compound isolated from Scutellaria radix. Accumulating evidence suggests that wogonin plays a crucial role in anti-inflammatory and anti-oxidative stress. Treatment of microglia with nuclear receptor agonists augments the expression of phagocytosis-related genes. However, the neuroprotective effects of wogonin in ICH remain obscure. In this study, we elucidated an innovative mechanism by which wogonin acts to enhance phagocytosis in a murine model of ICH. Wogonin promoted hematoma clearance and improved neurological recovery after ICH by upregulating the expression of Axl, MerTK, CD36, and LAMP2 in perihematomal microglia and BV2 cells. Treatment of a murine model of ICH with wogonin stimulated microglial phagocytosis in vitro. Further, we demonstrated that wogonin dramatically attenuated inflammatory and oxidative stress responses in a murine model of ICH by reducing the expression of pro-inflammatory cytokines and pro-oxidant enzymes such as TNF-α, IL-1β, and inducible nitric oxide synthase (iNOS) after ICH. The effects of wogonin were abolished by administration of the PPAR-γ inhibitor GW9662. In conclusion, our data suggest that wogonin facilitates hematoma clearance and neurobehavioral recovery by targeting PPAR-γ.
Collapse
|
19
|
Krenzlin H, Gresser E, Jussen D, Riede N, Taylor L, Vogelaar CF, Ringel F, Kempski O, Alessandri B. The Cerebral Thrombin System Is Activated after Intracerebral Hemorrhage and Contributes to Secondary Lesion Growth and Poor Neurological Outcome in C57Bl/6 Mice. J Neurotrauma 2020; 37:1481-1490. [PMID: 31830857 DOI: 10.1089/neu.2019.6582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
With increasing evidence for the existence of a cerebral thrombin system, coagulation factor IIa (thrombin) is suspected to influence the pathogenesis of secondary injury progression after intracerebral hemorrhage (ICH). We hypothesized that mechanisms associated with local volume expansion after ICH, rather than blood constituents, activate the cerebral thrombin system and are responsible for detrimental neurological outcome. To test this hypothesis, we examine the local thrombin expression after ICH in a C57BL/6N mouse model in the presence and absence of blood constituents. ICH was established using stereotaxic orthotopic injection of utologous blood (n = 10) or silicone oil as inert volume substance (n = 10) into the striatum. Intracranial pressure (ICP), cerebral blood flow (CBF), and mean arterial blood pressure (MAP) were monitored during and 30 min after the procedure. No significant differences between ICP, CBF, and MAP were found between both groups. Prothrombin messenger RNA expression was upregulated early after ICH. Immunohistochemistry showed an increase of perilesional thrombin in both groups (blood, 4.24-fold; silicone, 3.10-fold), whereas prothrombin fragment (F1.2) was elevated only in the absence of whole blood. Thrombin expression is colocalized with neuronal antigen expression. After 24 h, lesion size and neuronal loss were similar. Perihematomal thrombin correlated with increased neuronal loss and detrimental neurological outcome in vivo. In our study, we demonstrate, for the first time, that the local cerebral thrombin system is activated after ICH and that this activation is independent of the presence of whole-blood constituents. In our study, neuronal damage is driven by local thrombin expression and leads to an adverse clinical outcome.
Collapse
Affiliation(s)
- Harald Krenzlin
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany.,Department of Neurosurgery, and Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Eva Gresser
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Daniel Jussen
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany.,Department of Neurosurgery, HELIOS Dr. Horst-Schmidt-Kliniken, Wiesbaden, Germany
| | - Nicole Riede
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Louise Taylor
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | | | - Florian Ringel
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany.,Department of Neurosurgery, and Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Oliver Kempski
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Beat Alessandri
- Institute of Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
20
|
Wan Y, Gao F, Ye F, Yang W, Hua Y, Keep RF, Xi G. Effects of aging on hydrocephalus after intraventricular hemorrhage. Fluids Barriers CNS 2020; 17:8. [PMID: 32106865 PMCID: PMC7047364 DOI: 10.1186/s12987-020-0169-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hydrocephalus is a common and major complication that affects outcome after intraventricular hemorrhage (IVH). While aging impacts the occurrence of hydrocephalus in patients with IVH this and the underlying mechanisms have received little attention. The present investigation, therefore, studied the impact of aging on hydrocephalus after IVH in a rat model. METHODS Young and aged (3 and 18 months old, respectively) male Fischer 344 rats had an intraventricular injection of 200 μl autologous blood or saline. Ventricular volume was estimated using magnetic resonance imaging (MRI), while ventricular wall damage, heme oxygenase-1 (HO-1) and epiplexus cell activation were quantified by histological staining and Western blot. Additionally, the impact of intraventricular iron injection was examined in young and aged rats. RESULTS Intraventricular injection of autologous blood induced hydrocephalus in both young and aged rats but ventricular volumes were larger in aged rats compared to young rats from day 3 to day 14 followed IVH. In addition, ventricular wall damage and periventricular HO-1 upregulation were greater in aged versus young rats on day 1 after IVH. Aged rats also had more choroid plexus epiplexus cells on day 14 after IVH. Additionally, organized hematomas were observed in 23% (3/13) of aged rats but not in young rats after IVH. Organized hematomas in aged rats showed larger T2* lesions on MRI compared to rats with non-organized hematomas. Similar to the effects of IVH, intraventricular injection of iron resulted in more epiplexus cells activation and more severe hydrocephalus in aged compared to young rats. CONCLUSIONS IVH causes more severe hydrocephalus in aged compared to young rats. Enhanced ventricular wall damage, epiplexus cell activation and iron overload may contribute to this aggravated hydrocephalus development in aged animals.
Collapse
Affiliation(s)
- Yingfeng Wan
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Feng Gao
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- Department of Neurology, 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Weiming Yang
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
21
|
Bocheng W, Chaofeng L, Chuan C, Haiyong H, Tengchao H, Qun G, Ying G. Intracranial lymphatic drainage discharges iron from the ventricles and reduce the occurrence of chronic hydrocephalus after intraventricular hemorrhage in rats. Int J Neurosci 2019; 130:130-135. [PMID: 31516063 DOI: 10.1080/00207454.2019.1667780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wang Bocheng
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Liang Chaofeng
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chen Chuan
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - He Haiyong
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huang Tengchao
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Gao Qun
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guo Ying
- Neurosurgery department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
22
|
Wang M, Hua Y, Keep RF, Wan S, Novakovic N, Xi G. Complement Inhibition Attenuates Early Erythrolysis in the Hematoma and Brain Injury in Aged Rats. Stroke 2019; 50:1859-1868. [PMID: 31177985 DOI: 10.1161/strokeaha.119.025170] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background and Purpose- Early erythrolysis in the hematoma contributes to brain injury after intracerebral hemorrhage (ICH). This study investigated the effects of N-acetylheparin, a complement inhibitor, and aurin tricarboxylic acid, a membrane attack complex inhibitor, on early erythrolysis, brain iron deposition, and brain injury in aged rats. Methods- There were 3 parts in the study. First, aged (18 months old) male Fischer 344 rats had an ICH. The time course of erythrolysis in the hematoma was determined by T2* weighted magnetic resonance imaging, and the expression of CD163 was examined. Second, aged rats had an ICH with N-acetylheparin or vehicle. Rats were euthanized at days 1, 3, and 28 after magnetic resonance imaging (T2-, T2*-weighted, and T2* array) and behavioral tests. Brains were used for immunohistochemistry. Third, aged rats had an ICH with avaurin tricarboxylic acid or vehicle. The rats had magnetic resonance imaging and behavioral tests and were euthanized at day 3. Brains were used for immunohistochemistry. Results- Early erythrolysis occurred within the clot in aged F344 rats. There were increased numbers of CD163-positive cells after ICH. Almost all perihematomal CD163-positive cells were microglia/macrophages, while positive neurons were found more distant from the hematoma. Coinjection of N-acetylheparin attenuated erythrolysis, iron accumulation, CD163 expression, microglia activation, brain swelling, and neuronal death in the acute phase, as well as reducing brain atrophy and neurological deficits in the chronic phase. Coinjection of aurin tricarboxylic acid also reduced erythrolysis and ICH-induced brain injury. Conclusions- Inhibiting complement activation resulted in less erythrolysis and brain injury after ICH.
Collapse
Affiliation(s)
- Ming Wang
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., Y.H., R.F.K., S.W., N.N., G.X.).,Department of Neurosurgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (M.W.)
| | - Ya Hua
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., Y.H., R.F.K., S.W., N.N., G.X.)
| | - Richard F Keep
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., Y.H., R.F.K., S.W., N.N., G.X.)
| | - Shu Wan
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., Y.H., R.F.K., S.W., N.N., G.X.)
| | - Nemanja Novakovic
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., Y.H., R.F.K., S.W., N.N., G.X.)
| | - Guohua Xi
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., Y.H., R.F.K., S.W., N.N., G.X.)
| |
Collapse
|
23
|
Li F, Li X, Yang J, Guo X, Zheng X, Lv Z, Shi C. Increased Expression of Apo-J and Omi/HtrA2 After Intracerebral Hemorrhage in Rats. World Neurosurg 2018; 116:e26-e34. [PMID: 29581019 DOI: 10.1016/j.wneu.2018.03.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To investigate the changes of Apo-J and Omi/HtrA2 protein expression in rats with intracerebral hemorrhage. METHODS 150 Sprague-Dawley adult rats were randomly divided into 3 groups: (1) normal control (NC) group, (2) sham group, and (3) intracerebral hemorrhage (ICH) group. The data were collected at 6 hours, 12 hours, 1 day, 2 days, 3 days, 5 days, and 7 days. Apoptosis was measured by terminal deoxynucleotidyl transferase-mediated biotinylated-dUTP nick-end labeling staining. The distributions of the Apo-J and Omi/HtrA2 proteins were determined by immunohistochemical staining. The levels of Apo-J mRNA and Omi/HtrA2 mRNA expressions were examined by real-time polymerase chain reaction. RESULTS Apoptosis in the ICH group was higher than in the sham and NC groups (P < 0.05). Both the Apo-J and Omi/HtrA2 expression levels were increased in the peripheral region of hemorrhage, with a peak at 3 days. The Apo-J mRNA level positively correlated with the HtrA2 mRNA level in the ICH group (r = 0.883, P < 0.001). CONCLUSION The expressions of Apo-J and Omi/HtrA2 increased in parallel in the peripheral region of rat cerebral hemorrhage. Local high expression of Apo-J in the peripheral regions may play a neuroprotective role by inhibiting apoptosis via the Omi/HtrA2 pathway after hemorrhage.
Collapse
Affiliation(s)
- Feng Li
- Department of Neurology, Wenjiang District People's Hospital, Chengdu, China
| | - Xiaogang Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Yang
- Department of Neurology, Wenjiang District People's Hospital, Chengdu, China
| | - Xiaoyan Guo
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaomei Zheng
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhiyu Lv
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changqing Shi
- Department of Neurosurgery, Wenjiang District People's Hospital, Chengdu, China.
| |
Collapse
|
24
|
Chang CF, Goods BA, Askenase MH, Hammond MD, Renfroe SC, Steinschneider AF, Landreneau MJ, Ai Y, Beatty HE, da Costa LHA, Mack M, Sheth KN, Greer DM, Huttner A, Coman D, Hyder F, Ghosh S, Rothlin CV, Love JC, Sansing LH. Erythrocyte efferocytosis modulates macrophages towards recovery after intracerebral hemorrhage. J Clin Invest 2018; 128:607-624. [PMID: 29251628 PMCID: PMC5785262 DOI: 10.1172/jci95612] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/07/2017] [Indexed: 02/03/2023] Open
Abstract
Macrophages are a source of both proinflammatory and restorative functions in damaged tissue through complex dynamic phenotypic changes. Here, we sought to determine whether monocyte-derived macrophages (MDMs) contribute to recovery after acute sterile brain injury. By profiling the transcriptional dynamics of MDMs in the murine brain after experimental intracerebral hemorrhage (ICH), we found robust phenotypic changes in the infiltrating MDMs over time and demonstrated that MDMs are essential for optimal hematoma clearance and neurological recovery. Next, we identified the mechanism by which the engulfment of erythrocytes with exposed phosphatidylserine directly modulated the phenotype of both murine and human MDMs. In mice, loss of receptor tyrosine kinases AXL and MERTK reduced efferocytosis of eryptotic erythrocytes and hematoma clearance, worsened neurological recovery, exacerbated iron deposition, and decreased alternative activation of macrophages after ICH. Patients with higher circulating soluble AXL had poor 1-year outcomes after ICH onset, suggesting that therapeutically augmenting efferocytosis may improve functional outcomes by both reducing tissue injury and promoting the development of reparative macrophage responses. Thus, our results identify the efferocytosis of eryptotic erythrocytes through AXL/MERTK as a critical mechanism modulating macrophage phenotype and contributing to recovery from ICH.
Collapse
Affiliation(s)
- Che-Feng Chang
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Brittany A. Goods
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Michael H. Askenase
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Matthew D. Hammond
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephen C. Renfroe
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Margaret J. Landreneau
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Youxi Ai
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hannah E. Beatty
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Luís Henrique Angenendt da Costa
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Matthias Mack
- Department of Internal Medicine (Nephrology), University of Regensburg, Regensburg, Germany
| | - Kevin N. Sheth
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - David M. Greer
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Daniel Coman
- Department of Diagnostic Radiology and Biomedical Engineering
| | - Fahmeed Hyder
- Department of Diagnostic Radiology and Biomedical Engineering
| | - Sourav Ghosh
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology, and
| | - Carla V. Rothlin
- Department of Pharmacology, and
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - J. Christopher Love
- Chemical Engineering, Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
25
|
Hirt L, Price M, Mastour N, Brunet JF, Barrière G, Friscourt F, Badaut J. Increase of aquaporin 9 expression in astrocytes participates in astrogliosis. J Neurosci Res 2017; 96:194-206. [PMID: 28419510 DOI: 10.1002/jnr.24061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 01/01/2023]
Abstract
Here we assess the potential functional role of increased aquaporin 9 (APQ9) in astrocytes. Increased AQP9 expression was achieved in primary astrocyte cultures by transfection of a plasmid-containing green fluorescent protein fused to either wild-type or mutated human AQP9. Increased AQP9 expression and phosphorylation at Ser222 were associated with a significant change in astrocyte morphology, mainly with a higher number of processes. Similar phenotypic changes are observed in astrogliosis processes after injury. In parallel, we observed that in vivo, thrombin preconditioning before ischemic stroke induced an early increase in AQP9 expression in the male mouse brain. This increased AQP9 expression was also associated with astrocyte morphological changes, especially in the white matter tract. Astrocyte reactivity is debated as being either beneficial or deleterious. As thrombin preconditioning leads to a decrease in lesion size after stroke, our data suggest that the early increase in AQP9 concomitant with astrocyte reactivity leads to a beneficial effect. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lorenz Hirt
- Neurology Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Melanie Price
- Neurology Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Nabil Mastour
- Neurosurgery Research Group, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Jean-François Brunet
- Neurosurgery Research Group, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | - Jerome Badaut
- CNRS UMR 5287, INCIA, University of Bordeaux, Bordeaux, France
| |
Collapse
|
26
|
Lim-Hing K, Rincon F. Secondary Hematoma Expansion and Perihemorrhagic Edema after Intracerebral Hemorrhage: From Bench Work to Practical Aspects. Front Neurol 2017; 8:74. [PMID: 28439253 PMCID: PMC5383656 DOI: 10.3389/fneur.2017.00074] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 02/20/2017] [Indexed: 01/24/2023] Open
Abstract
Intracerebral hemorrhages (ICH) represent about 10-15% of all strokes per year in the United States alone. Key variables influencing the long-term outcome after ICH are hematoma size and growth. Although death may occur at the time of the hemorrhage, delayed neurologic deterioration frequently occurs with hematoma growth and neuronal injury of the surrounding tissue. Perihematoma edema has also been implicated as a contributing factor for delayed neurologic deterioration after ICH. Cerebral edema results from both blood-brain barrier disruption and local generation of osmotically active substances. Inflammatory cellular mediators, activation of the complement, by-products of coagulation and hemolysis such as thrombin and fibrin, and hemoglobin enter the brain and induce a local and systemic inflammatory reaction. These complex cascades lead to apoptosis or neuronal injury. By identifying the major modulators of cerebral edema after ICH, a therapeutic target to counter degenerative events may be forthcoming.
Collapse
Affiliation(s)
- Krista Lim-Hing
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Fred Rincon
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
27
|
Bao L, Zu J, He Q, Zhao H, Zhou S, Ye X, Yang X, Zan K, Zhang Z, Shi H, Cui G. Thrombin-induced apoptosis in neurons through activation of c-Jun-N-terminal kinase. Toxicol Mech Methods 2016; 27:18-23. [PMID: 27841083 DOI: 10.3109/15376516.2016.1172691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Studies have shown that thrombin activation played a central role in cell injuries associated with intracerebral hemorrhage (ICH). OBJECTIVE Here, our study investigated the cytotoxicity of thrombin on neurons, and determined the involvement of JNK pathways in thrombin-induced neuronal apoptosis. MATERIALS AND METHODS Primary cultured neurons were treated with different doses of thrombin. Some neurons were given either SP600125 or vehicle. LDH release assay and flow cytometry were used to measure neuronal apoptosis caused by thrombin. The activation of JNK and capases-3 were measured by Western blot. RESULTS Our results showed large doses of thrombin that increased the LDH release, the level of cleaved caspase-3 and apoptosis rate of neurons. JNK was activated by thrombin in a time-dependent manner. Administration of SP600125 protects neurons from thrombin-induced apoptosis. CONCLUSION These data indicate that the activation of JNK is crucial for thrombin-induced neuronal apoptosis, and inhibition of JNK may be a potential therapeutic target for ICH.
Collapse
Affiliation(s)
- Lei Bao
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Jie Zu
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Qianqian He
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Hui Zhao
- b Department of Neurology , Xuzhou Central Hospital , Xuzhou , Jiangsu , China
| | - Su Zhou
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Xinchun Ye
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Xinxin Yang
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Kun Zan
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Zuohui Zhang
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Hongjuan Shi
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Guiyun Cui
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| |
Collapse
|
28
|
Guan J, Zhang S, Zhou Q, Yuan Z, Lu Z. Effect of thrombin preconditioning on migration of subventricular zone-derived cells after intracerebral hemorrhage in rats. Neurol Res 2016; 38:809-16. [PMID: 27477964 DOI: 10.1080/01616412.2016.1210356] [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: 01/19/2023]
Abstract
OBJECTIVE To investigate the effect of thrombin preconditioning (TPC) on the intracerebral hemorrhage (ICH)-induced proliferation, migration, and function of subventriclular zone (SVZ) cells and to find new strategies that enhance endogenous neurogenesis after ICH. METHODS Male Sprague-Dawley rats were randomly divided into 3 groups (ICH, TPC, and control group). Rats of each group were randomly divided into 5 subgroups (3-d, 7-d, 14-d, 21-d, and 28-d subgroup). ICH was caused by intrastrial stereotactic administration of collagenase type IV. Brdu was used to label newborn SVZ cells. Organotypic brain slices were cultured to dynamically observe the migration of SVZ cells at living brain tissue. Migration of Dil-labeled SVZ cells in living brain slices was traced by time-lapse microscopy. To assess whether SVZ cells migrating to injured striatum had the ability to form synapses with other cells, brain slices from each group were double immunolabeled with Brdu and synapsin I. RESULTS The number of Brdu-positive cells markedly increased in the ipsilateral SVZ and striatum 3 days after TPC, peaked at 14 days (P < 0.01), continued to 21 days, and then gradually decreased at 28 days with significant difference compared to the ICH group at each time point (P < 0.01). Migration of Dil-labeled SVZ cells in brain slices in each group was observed and imaged during a 12-h period. Dil-labeled SVZ cells in the TPC group were observed to migrate laterally toward striatum with time with a faster velocity compared to the ICH group (P < 0.01). Our study also demonstrated that TPC induced strong colocalization of Brdu and synapsin I in the ipsilateral striatum between 3 and 28 days after injury.TPC made colocalization of Brdu and synapsin I appear earlier and continue for a longer time compared to the ICH group. CONCLUSIONS Our results demonstrated that TPC could promote proliferation, migration, and function of SVZ cells after ICH, which may provide a new idea for enhancing endogenous neurogenesis and developing new therapeutic strategies against ICH-induced brain injury.
Collapse
Affiliation(s)
- Jingxia Guan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Shaofeng Zhang
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Qin Zhou
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zhenhua Yuan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zuneng Lu
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| |
Collapse
|
29
|
Rohatgi T, Sedehizade F, Reymann KG, Reiser G. Protease-Activated Receptors in Neuronal Development, Neurodegeneration, and Neuroprotection: Thrombin as Signaling Molecule in the Brain. Neuroscientist 2016; 10:501-12. [PMID: 15534036 DOI: 10.1177/1073858404269955] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protease-activated receptors (PARs) belong to the superfamily of seven transmembrane domain G protein-coupled receptors. Four PAR subtypes are known, PAR-1 to -4. PARs are highly homologous between the species and are expressed in a wide variety of tissues and cell types. Of particular interest is the role which these receptors play in the brain, with regard to neuroprotection or degeneration under pathological conditions. The main agonist of PARs is thrombin, a multifunctional serine protease, known to be present not only in blood plasma but also in the brain. PARs possess an irreversible activation mechanism. Binding of agonist and subsequent cleavage of the extracellular N-terminus of the receptor results in exposure of a so-called tethered ligand domain, which then binds to extracellular loop 2 of the receptor leading to receptor activation. PARs exhibit an extensive expression pattern in both the central and the peripheral nervous system. PARs participate in several mechanisms important for normal cellular functioning and during critical situations involving cellular survival and death. In the last few years, research on Alzheimer’s disease and stroke has linked PARs to the pathophysiology of these neurodegenerative disorders. Actions of thrombin are concentration-dependent, and therefore, depending on cellular function and environment, serve as a double-edged sword. Thrombin can be neuroprotective during stress conditions, whereas under normal conditions high concentrations of thrombin are toxic to cells.
Collapse
Affiliation(s)
- Tanuja Rohatgi
- Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Magdeburg, Germany
| | | | | | | |
Collapse
|
30
|
Wan S, Cheng Y, Jin H, Guo D, Hua Y, Keep RF, Xi G. Microglia Activation and Polarization After Intracerebral Hemorrhage in Mice: the Role of Protease-Activated Receptor-1. Transl Stroke Res 2016; 7:478-487. [PMID: 27206851 DOI: 10.1007/s12975-016-0472-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 02/06/2023]
Abstract
Polarized microglia play a dual (beneficial/detrimental) role in neurological diseases. However, the status and the factors that modulate microglia polarization in intracerebral hemorrhage (ICH) remain unclear. In the present study, we investigated the role of protease-activated receptor-1 (PAR-1, a thrombin receptor) in ICH-induced microglia polarization in mice. Male wild-type (WT) and PAR-1 knockout (PAR-1 KO) mice received an infusion of 30-μL autologous blood or saline into the right basal ganglia. Mice were euthanized at different time points and the brains were used for Western blotting and immunohistochemistry. Some mice had magnetic resonance imaging. We found that ICH induced microglia activation and polarization. M1 phenotypic markers were markedly increased and reached a peak as early as 4 h, remained high at 3 days and decreased 7 days after ICH. M2 phenotypic markers were upregulated later than M1 markers reaching a peak at day 1 and declining by day 7 after ICH. PAR-1 was upregulated after ICH and expressed in the neurons and microglia. ICH induced less brain swelling and neuronal death in PAR-1 KO mice, and this was associated with less M1 polarization and reduced proinflammatory cytokine levels in the brain. In conclusion, these results suggest that polarized microglia occur dynamically after ICH and that PAR-1 plays a role in the microglia activation and polarization.
Collapse
Affiliation(s)
- Shu Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingying Cheng
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurology, The 1st Affiliated Hospital, School of Medicine, Jilin University, Changchun, China
| | - Hang Jin
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurology, The 1st Affiliated Hospital, School of Medicine, Jilin University, Changchun, China
| | - Dewei Guo
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA. .,University of Michigan, Room5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
31
|
Mao S, Xi G, Keep RF, Hua Y. Role of Lipocalin-2 in Thrombin-Induced Brain Injury. Stroke 2016; 47:1078-84. [PMID: 26869387 DOI: 10.1161/strokeaha.115.012153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Thrombin and lipocalin-2 (LCN2) contribute to intracerebral hemorrhage-induced brain injury. Thrombin-induced brain damage is partially through a thrombin receptor, protease-activated receptor-1. LCN2 is involved in cellular iron transport and neuroinflammation. This study investigated the role of LCN2 in thrombin-induced brain injury. METHODS There were 3 parts in this study. First, male adult C57BL/6 wild-type or LCN2 knockout (LCN2 KO) mice had an intracaudate injection of thrombin (0.4 U) or saline. Second, LCN2 KO mice had an injection of thrombin (0.4 U) with recombinant mouse LCN2 protein (1 μg) into the right caudate. Third, protease-activated receptor-1 KO or wild-type mice had an intracaudate injection of thrombin or saline. All mice had T2-weighted magnetic resonance imaging and behavioral tests. Brains were used for histology, immunohistochemistry, and Western blotting. RESULTS Intracerebral thrombin injection caused LCN2 upregulation and brain injury in mice. Thrombin-induced brain swelling, blood-brain barrier disruption, neuronal death, and neurological deficits were markedly less in LCN2 KO mice (P<0.05) and were exacerbated by exogenous LCN2 coinjection. In addition, thrombin injection resulted in less LCN2 expression and brain injury in protease-activated receptor-1 KO mice. CONCLUSIONS Thrombin upregulates LCN2 through protease-activated receptor-1 activation and causes brain damage.
Collapse
Affiliation(s)
- Shanshan Mao
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Guohua Xi
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Richard F Keep
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Ya Hua
- From the Department of Neurosurgery, University of Michigan, Ann Arbor.
| |
Collapse
|
32
|
The Importance of Thrombin in Cerebral Injury and Disease. Int J Mol Sci 2016; 17:ijms17010084. [PMID: 26761005 PMCID: PMC4730327 DOI: 10.3390/ijms17010084] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/28/2015] [Accepted: 12/30/2015] [Indexed: 12/31/2022] Open
Abstract
There is increasing evidence that prothrombin and its active derivative thrombin are expressed locally in the central nervous system. So far, little is known about the physiological and pathophysiological functions exerted by thrombin in the human brain. Extra-hepatic prothrombin expression has been identified in neuronal cells and astrocytes via mRNA measurement. The actual amount of brain derived prothrombin is expected to be 1% or less compared to that in the liver. The role in brain injury depends upon its concentration, as higher amounts cause neuroinflammation and apoptosis, while lower concentrations might even be cytoprotective. Its involvement in numerous diseases like Alzheimer’s, multiple sclerosis, cerebral ischemia and haemorrhage is becoming increasingly clear. This review focuses on elucidation of the cerebral thrombin expression, local generation and its role in injury and disease of the central nervous system.
Collapse
|
33
|
Ni W, Mao S, Xi G, Keep RF, Hua Y. Role of Erythrocyte CD47 in Intracerebral Hematoma Clearance. Stroke 2016; 47:505-11. [PMID: 26732568 DOI: 10.1161/strokeaha.115.010920] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/01/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE Enhancing hematoma clearance through phagocytosis may reduce brain injury after intracerebral hemorrhage. In the current study, we investigated the role of cluster of differentiation 47 (CD47) in regulating erythrophagocytosis and brain injury after intracerebral hemorrhage in nude mice. METHODS This study was in 2 parts. First, male adult nude mice had an intracaudate injection of 30 μL saline, blood from male adult wild-type (WT) mice, or blood from CD47 knockout mice. Second, mice had an intracaudate injection of 30 μL CD47 knockout blood with clodronate or control liposomes. Clodronate liposomes were also tested in saline-injected mice. All mice then had magnetic resonance imaging to measure hematoma size and brain swelling. Brains were used for immunohistochemistry and Western blot. RESULTS Erythrophagocytosis occurred in and around the hematoma. Injection of CD47 knockout blood resulted in quicker clot resolution, less brain swelling, and less neurological deficits compared with wild-type blood. Higher brain heme oxygenase-1 levels and more microglial activation (mostly M2 polarized microglia) at day 3 were found after CD47 knockout blood injection. Co-injection of clodronate liposomes, to deplete phagocytes, caused more severe brain swelling and less clot resolution. CONCLUSIONS These results indicated that CD47 has a key role in hematoma clearance after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Wei Ni
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Shanshan Mao
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Guohua Xi
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Richard F Keep
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Ya Hua
- From the Department of Neurosurgery, University of Michigan, Ann Arbor.
| |
Collapse
|
34
|
Zhao J, Xi G, Wu G, Keep RF, Hua Y. Deferoxamine Attenuated the Upregulation of Lipocalin-2 Induced by Traumatic Brain Injury in Rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:291-4. [PMID: 26463963 DOI: 10.1007/978-3-319-18497-5_50] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intracranial hemorrhage is one of the most common consequences of traumatic brain injury (TBI). The release of iron from the breakdown of hemoglobin during intracerebral hematoma resolution results in an increase in perihematomal non-heme iron. Lipocalin 2 (LCN-2) is a siderophore-binding protein that mediates transferrin-independent iron transport. This study examined the effects of TBI (lateral fluid percussion) on LCN-2 expression in Sprague-Dawley rats. LCN-2 protein levels were markedly increased in the ipsilateral cortex and hippocampus after TBI, with the highest level at day 1. Most LCN-2-positive cells appeared to be astrocytes. Treatment with an iron chelator, deferoxamine (100 mg/kg, intramuscularly), attenuated the TBI-induced upregulation of LCN-2. In summary, TBI resulted in upregulation of LCN-2 and deferoxamine reduced TBI-induced LCN-2 increase, suggesting LCN-2 may have a role in iron-trafficking after TBI.
Collapse
Affiliation(s)
- Jinbing Zhao
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Gang Wu
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
35
|
Benggon M, Chen H, Applegate RL, Zhang J. Thrombin Preconditioning in Surgical Brain Injury in Rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:299-304. [PMID: 26463965 DOI: 10.1007/978-3-319-18497-5_52] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The surgical brain injury model replicates neurosurgical brain parenchymal damage. Postsurgical brain edema correlates with postoperative neurological dysfunction. Intranasal administration is a proven method of delivering therapies to brain tissue. Thrombin preconditioning decreased brain edema and improved neurological outcomes in models of ischemic brain injury. We hypothesized thrombin preconditioning in surgical brain injury may improve postoperative brain edema and neurological outcomes. Adult male Sprague-Dawley rats (n = 78) weighing 285-355 g were randomly assigned to sham or pre-injury treatment: one-time pretreatment 1 day prior, one-time pretreatment 5 days prior, and daily preconditioning for 5 days prior. Treatment arms were divided into vehicle or thrombin therapies, and subdivided into intranasal (thrombin 5 units/50 μL 0.9 % saline) or intracerebral ventricular (thrombin 0.1 unit/10 μL 0.9 % saline) administration. Blinded observers performed neurological testing 24 h after brain injury followed immediately by measurement of brain water content. There was a significant difference in ipsilateral brain water content and neurological outcomes between all treatment groups and the sham group. However, there was no change in brain water content or neurological outcomes between thrombin- and vehicle-treated animals. Thrombin preconditioning did not significantly improve brain edema or neurological function in surgical brain injury in rats.
Collapse
Affiliation(s)
- Michael Benggon
- Department of Anesthesiology, Loma Linda University, School of Medicine, 11234 Anderson Street, Loma Linda, CA, USA
| | - Hank Chen
- Department of Basic Science, Division of Physiology, Loma Linda School of Medicine, Loma Linda, CA, USA
| | - Richard L Applegate
- Department of Anesthesiology, Loma Linda University, School of Medicine, Room 2532 LLUMC, 11234 Anderson Street, Loma Linda, CA, 92374, USA.
| | - John Zhang
- Department of Basic Science, Division of Physiology, Loma Linda School of Medicine, Loma Linda, CA, USA
- Department of Anesthesiology, Loma Linda University, School of Medicine, Room 2532 LLUMC, 11234 Anderson Street, Loma Linda, CA, 92374, USA
| |
Collapse
|
36
|
Effect of Gender on Iron-induced Brain Injury in Low Aerobic Capacity Rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:367-71. [PMID: 26463976 DOI: 10.1007/978-3-319-18497-5_63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain iron overload has a key role in brain injury after intracerebral hemorrhage (ICH). Low aerobic capacity is a risk factor for cardiovascular disease and our previous study demonstrated that ICH-induced brain injury is enhanced in rats with low aerobic capacity (low capacity runners; LCRs). We have found that ICH-induced injury is less in female rats compared with that in males. In the present study, we examined the effects of gender on iron-induced brain injury in rats with low aerobic capacity. Adult male and female LCR rats had an intracaudate injection of FeCl2 (50 μl 0.5 mM). T2 Magnetic resonance imaging was carried out at 24 h to determine brain swelling and T2 brain lesion volume. Albumin leakage, an indicator of blood-brain barrier (BBB) disruption, and heme oxygenase-1 (HO-1, a stress marker) levels were determined. Male LCR rats had more severe hemisphere swelling (difference of ipsilateral to contralateral hemisphere volume: 16.6 ± 4.1 vs 11.1 ± 2.6 % in females, p < 0.05) and larger T2 lesion volumes (120 ± 28 vs 87 ± 27 mm(3) in females, p < 0.05) after iron injection. Iron also resulted in more severe BBB disruption in the ipsilateral hemisphere of males (albumin levels: 7,717 ± 1,502 pixels in males vs 5,287 ± 1,342 pixels in females; p < 0.05). The immunoreactivity of HO-1 was also significantly higher in males than females (HO-1/β-actin: 1.31 ± 0.44 vs 1.03 ± 0.05, p < 0.05). Female LCR rats had less iron-induced brain swelling, smaller lesion volumes, and reduced BBB disruption and HO-1 upregulation compared with male LCR rats. This may contribute to the reduced ICH-induced brain injury found in females.
Collapse
|
37
|
Effects of Aerobic Capacity on Thrombin-Induced Hydrocephalus and White Matter Injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:379-84. [PMID: 26463978 DOI: 10.1007/978-3-319-18497-5_65] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
We have previously shown that intracerebral hemorrhage-induced brain injury is less in rats bred for high aerobic capacity (high capacity runners; HCR) compared with those bred for low aerobic capacity (low capacity runners; LCRs). Thrombin, an essential component in the coagulation cascade, is produced after cerebral hemorrhage. Intraventricular injection of thrombin causes significant hydrocephalus and white matter damage. In the present study, we examined the effect of exercise capacity on thrombin-induced hydrocephalus and white matter damage. Mid-aged (13-month-old) female LCRs (n = 13) and HCRs (n = 12) rats were used in this study. Rats received an intraventricular injection of thrombin (3 U, 50 μl). All rats underwent magnetic resonance imaging (MRI) at 24 h and were then euthanized for brain histology and Western blot. The mortalities were 20 % in LCRs and 33 % in HCRs after thrombin injection (p > 0.05). No rats died after saline injection. Intraventricular thrombin injection resulted in hydrocephalus and periventricular white matter damage as determined on MRI. In LCR rats, thrombin induced significant ventricle enlargement (23.0 ± 2.3 vs12.8 ± 1.9 mm(3) in LCR saline group; p < 0.01) and white matter lesion (9.3 ± 7.6 vs 0.6 ± 0.5 mm(3) in LCR saline group, p < 0.05). In comparison, in HCR rats thrombin induced less ventricular enlargement (17.3 ± 3.9 vs 23.0 ± 2.3 mm(3) in LCRs, p < 0.01) and smaller white matter lesions (2.6 ± 1.2 mm(3) vs 9.3 ± 7.6 mm(3) in LCRs, p < 0.05). In LCR rats, there was also upregulation of heat shock protein-32, a stress marker, and microglial activation in the periventricular white matter. These changes were significantly reduced in HCR rats. Intraventricular injection of thrombin caused more white matter damage and hydrocephalus in rats with low aerobic capacity. A differential effect of thrombin may contribute to differences in the effects of cerebral hemorrhage with aerobic capacity.
Collapse
|
38
|
Li L, Tao Y, Tang J, Chen Q, Yang Y, Feng Z, Chen Y, Yang L, Yang Y, Zhu G, Feng H, Chen Z. A Cannabinoid Receptor 2 Agonist Prevents Thrombin-Induced Blood-Brain Barrier Damage via the Inhibition of Microglial Activation and Matrix Metalloproteinase Expression in Rats. Transl Stroke Res 2015; 6:467-77. [PMID: 26376816 DOI: 10.1007/s12975-015-0425-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 08/31/2015] [Accepted: 09/02/2015] [Indexed: 11/26/2022]
Abstract
Thrombin mediates the life-threatening cerebral edema and blood-brain barrier (BBB) damage that occurs after intracerebral hemorrhage (ICH). We previously found that the selective cannabinoid receptor 2 (CB2R) agonist JWH-133 reduced brain edema and neurological deficits following germinal matrix hemorrhage (GMH). We explored whether CB2R stimulation ameliorated thrombin-induced brain edema and BBB permeability as well as the possible molecular mechanism involved. A total of 144 Sprague-Dawley (S-D) rats received a thrombin (20 U) injection in the right basal ganglia. JWH-133 (1.5 mg/kg) or SR-144528 (3.0 mg/kg) and vehicle were intraperitoneally (i.p.) injected 1 h after surgery. Brain water content measurement, Evans blue (EB) extravasation, Western blot, and immunofluorescence were used to study the effects of a CB2R agonist 24 h after surgery. The results demonstrated that JWH-133 administration significantly decreased thrombin-induced brain edema and reduced the number of Iba-1-positive microglia. JWH-133 also decreased the number of P44/P42(+)/Iba-1(+) microglia, lowered Evans blue extravasation, and inhibited the elevated matrix metallopeptidase (MMP)-9 and matrix metallopeptidase (MMP)-12 activities. However, a selective CB2R antagonist (SR-144528) reversed these effects. We demonstrated that CB2R stimulation reduced thrombin-induced brain edema and alleviated BBB damage. We also found that matrix metalloproteinase suppression may be partially involved in these processes.
Collapse
Affiliation(s)
- Lin Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Yihao Tao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Zhou Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Liming Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Yunfeng Yang
- Department of Neurosurgery, Sichuan Provincial Corps Hospital, Chinese People's Armed Police Forces, Leshan, People's Republic of China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
39
|
Role of lipocalin-2 in brain injury after intracerebral hemorrhage. J Cereb Blood Flow Metab 2015; 35:1454-61. [PMID: 25853903 PMCID: PMC4640334 DOI: 10.1038/jcbfm.2015.52] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/24/2015] [Accepted: 03/02/2015] [Indexed: 01/08/2023]
Abstract
Lipocalin-2 (LCN2) is a siderophore-binding protein involved in cellular iron transport and neuroinflammation. Both iron and inflammation are involved in brain injury after intracerebral hemorrhage (ICH) and this study examined the role of LCN2 in such injury. Male adult C57BL/6 wild-type (WT) or LCN2-deficient (LCN2(-/-)) mice had an intracerebral injection of autologous blood or FeCl2. Control animals had a sham operation or saline injection. T2-weighted magnetic resonance imaging and behavioral tests were performed at days 1, 3, 7, 14, and 28 after injection. In WT mice, brain LCN2 levels were increased in the ipsilateral basal ganglia after ICH or iron injection. Lipocalin-2-positive cells were astrocytes, microglia, neurons, and endothelial cells. Intracerebral hemorrhage resulted in a significant increase in ferritin expression in the ipsilateral basal ganglia. Compared with WT mice, ICH caused less ferritin upregulation, microglia activation, brain swelling, brain atrophy, and neurologic deficits in LCN2(-/-) mice (P<0.05). The size of the lesion induced by FeCl2 injection as well as the degree of brain swelling and blood-brain barrier disruption were also less in LCN2(-/-) mice (P<0.05). These results suggest a role of LCN2 in enhancing brain injury and iron toxicity after ICH.
Collapse
|
40
|
Strahle JM, Garton T, Bazzi AA, Kilaru H, Garton HJL, Maher CO, Muraszko KM, Keep RF, Xi G. Role of hemoglobin and iron in hydrocephalus after neonatal intraventricular hemorrhage. Neurosurgery 2015; 75:696-705; discussion 706. [PMID: 25121790 DOI: 10.1227/neu.0000000000000524] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Neonatal germinal matrix hemorrhage/intraventricular hemorrhage is common and often results in hydrocephalus. The pathogenesis of posthemorrhagic hydrocephalus is not fully understood. OBJECTIVE To explore the potential role of hemoglobin and iron released after hemorrhage. METHODS Artificial cerebrospinal fluid (aCSF), hemoglobin, or iron was injected into the right lateral ventricle of postnatal day-7 Sprague Dawley rats. Ventricle size, heme oxygenase-1 (HO-1) expression, and the presence of iron were evaluated 24 and 72 hours after injection. A subset of animals was treated with an iron chelator (deferoxamine) or vehicle for 24 hours after hemoglobin injection, and ventricle size and cell death were evaluated. RESULTS Intraventricular injection of hemoglobin and iron resulted in ventricular enlargement at 24 hours compared with the injection of aCSF. Protoporphyrin IX, the iron-deficient immediate heme precursor, did not result in ventricular enlargement after injection into the ventricle. HO-1, the enzyme that releases iron from heme, was increased in the hippocampus and cortex of hemoglobin-injected animals at 24 hours compared with aCSF-injected controls. Treatment with an iron chelator, deferoxamine, decreased hemoglobin-induced ventricular enlargement and cell death. CONCLUSION Intraventricular injection of hemoglobin and iron can induce hydrocephalus. Treatment with an iron chelator reduced hemoglobin-induced ventricular enlargement. This has implications for the pathogenesis and treatment of posthemorrhagic hydrocephalus. ABBREVIATIONS aCSF, artificial cerebrospinal fluidDAB, 3,3'-diaminobenzidine-4HClGMH-IVH, germinal matrix hemorrhage/intraventricular hemorrhageHO-1, heme oxygenase-1ICH, intracerebral hemorrhagePBS, phosphate-buffered salineSVZ, subventricular zoneTBST, tris-buffered saline with Tween 20.
Collapse
|
41
|
Egashira Y, Hua Y, Keep RF, Xi G. Intercellular cross-talk in intracerebral hemorrhage. Brain Res 2015; 1623:97-109. [PMID: 25863131 DOI: 10.1016/j.brainres.2015.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disorder with high mortality and morbidity. Currently, there are few treatment strategies for ICH-induced brain injury. A recent increase in interest in the pathophysiology of ICH has led to elucidation of the pathways underlying ICH-induced brain injury, pathways where intercellular and hematoma to cell signaling play important roles. In this review, we summarize recent advances in ICH research focusing on intercellular and hematoma:cell cross-talk related to brain injury and recovery after ICH. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yusuke Egashira
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - 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
|
42
|
Correlating Cerebral (18)FDG PET-CT Patterns with Histological Analysis During Early Brain Injury in a Rat Subarachnoid Hemorrhage Model. Transl Stroke Res 2015; 6:290-5. [PMID: 25833084 DOI: 10.1007/s12975-015-0396-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 01/25/2023]
Abstract
Early brain injury (EBI) plays a significant role in poor outcomes for subarachnoid hemorrhage (SAH) patients. Further investigations are required to characterize the cellular metabolic and related histological changes that may contribute to EBI following SAH. We investigated the image patterns of 18-fluorodeoxyglucose positron emission tomography-computed tomography ((18)FDG PET-CT) during EBI and correlated histopathological changes utilizing a rat SAH model. SAH was induced in six adult male Sprague-Dawley rats by endovascular perforation, and animals were randomly assigned to receive (18)FDG PET-CT imaging at either 3 or 12 h post-procedure. Mean (18)FDG standard uptake value (SUV) of the brain was calculated. Animals were euthanized 48 h post-procedure, and brain samples were used for heme oxygenase-1 (HO-1) and dopamine- and cAMP-regulated phosphoprotein (DARPP-32) Mr 32 kDa immunohistochemistry. Rats within the SAH group had higher mean whole brain (18)FDG SUV (2.349 ± 0.376 g/ml in the 3-h group and 2.453 ± 0.495 g/ml in the 12-h group) compared to that of sham (n = 3; mean SUV = 2.030 ± 0.247 g/ml; P < 0.05) or control groups (n = 3; mean SUV = 1.800 ± 0.484 g/ml; P < 0.05). Whole brain (18)FDG SUV did not vary significantly between rats imaged at 3 h vs. those imaged at 12 h post-SAH (P > 0.05). Regions of decreasing SUV in SAH rats correlated with neuronal death and increased expression of HO-1. Higher (18)FDG PET SUV was evident in rats post-SAH compared to sham and control groups. Regions of decreasing SUV in SAH rats correlated with neuronal death and increased HO-1 expression as evaluated by histopathology.
Collapse
|
43
|
Zheng M, Du H, Ni W, Koch LG, Britton SL, Keep RF, Xi G, Hua Y. Iron-induced necrotic brain cell death in rats with different aerobic capacity. Transl Stroke Res 2015; 6:215-23. [PMID: 25649272 DOI: 10.1007/s12975-015-0388-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/16/2015] [Accepted: 01/21/2015] [Indexed: 12/29/2022]
Abstract
Brain iron overload has a key role in brain injury after intracerebral hemorrhage (ICH). Our recent study demonstrated that ICH-induced brain injury was greater in low capacity runner (LCR) than in high capacity runner (HCR) rats. The present study examines whether iron-induced brain injury differs between LCRs and HCRs. Adult male LCR and HCR rats had an intracaudate injection of iron or saline. Rats were euthanized at 2 and at 24 h after T2 magnetic resonance imaging, and the brains were used for immunostaining and Western blotting. LCRs had more hemispheric swelling, T2 lesion volumes, blood-brain barrier disruption, and neuronal death at 24 h after iron injection (p < 0.05). Many propidium iodide (PI)-positive cells, indicative of necrotic cell death, were observed in the ipsilateral basal ganglia of both HCRs and LCRs at 2 h after iron injection. PI fluorescence intensity was higher in LCRs than in HCRs. In addition, membrane attack complex (MAC) expression was increased at 2 h after iron injection and was higher in LCRs than in HCRs. The PI-positive cells co-localized with MAC-positive cells in the ipsilateral basal ganglia. Iron induces more severe necrotic brain cell death, brain swelling, and blood-brain barrier disruption in LCR rats, which may be related with complement activation and MAC formation.
Collapse
Affiliation(s)
- Mingzhe Zheng
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Merali Z, Leung J, Mikulis D, Silver F, Kassner A. Longitudinal assessment of imatinib's effect on the blood-brain barrier after ischemia/reperfusion injury with permeability MRI. Transl Stroke Res 2015; 6:39-49. [PMID: 25146090 DOI: 10.1007/s12975-014-0358-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/23/2014] [Accepted: 07/03/2014] [Indexed: 12/22/2022]
Abstract
Acute ischemic stroke (AIS) often results in degeneration of the blood-brain barrier (BBB), which can lead to vasogenic edema and an increased risk of intracerebral hemorrhage. Imatinib is an agent that may be able to protect the BBB and reduce the risk of the harmful consequences of BBB degeneration. We sought to measure the effect of Imatinib on the BBB after experimental stroke longitudinally in vivo with permeability dynamic contrast-enhanced MRI. Ischemia/reperfusion injury was induced with a transient middle cerebral artery occlusion surgery. Rats were given Imatinib at 2 and 20 h after stroke onset. Post-assessment included neurologic functioning, MR imaging, Evans Blue extravasation, Western blot, and immunohistology assay. Imatinib protected the BBB by 24 h but failed to decrease BBB permeability at an earlier time-point. Imatinib also reduced infarct volume, edema, and improved neurologic functioning by 24 h. Rats treated with Imatinib also had a higher expression of the BBB structural protein Zona ocludens-1 and a reduction in nuclear factor-kappa beta (NF-κβ) activation. Imatinib is a promising agent to protect the BBB after AIS, but its effect on the BBB may not become prominent until 24 h after the onset of ischemia. This finding may help elucidate Imatinib's role in the clinical management of AIS and influence future study designs.
Collapse
Affiliation(s)
- Zamir Merali
- Department of Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
45
|
Zhou QB, Jin YL, Jia Q, Zhang Y, Li LY, Liu P, Liu YT. Baicalin attenuates brain edema in a rat model of intracerebral hemorrhage. Inflammation 2014; 37:107-15. [PMID: 23974988 PMCID: PMC3929027 DOI: 10.1007/s10753-013-9717-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Baicalin is a flavonoid compound purified from the roots of Scutellaria baicalensis, which possesses multiple biological activities. Previous studies have shown that baicalin is protective in ischemic cerebral diseases. The aim of the present study was to examine the effects of baicalin on brain injury in a rat model of intracerebral hemorrhage (ICH) and to explore the possible mechanisms. Intracerebral hemorrhage was induced in male Wistar rats by injection of 0.5 U collagenaseVII to the caudate nucleus. Sham operation rats were injected with equal volume of saline. After the induction of ICH, the rats were randomly divided into four groups and administered with different dose of baicalin (0, 25, 50, or 100 mg/kg in saline) through peritoneal injection. The brain tissues around the hemorrhage areas were collected on days 1, 3, and 5 after treatment. Brain edema was analyzed by desiccation method; the metalloproteinase-9 (MMP-9) protein and mRNA expression were determined by western blotting and real time RT-PCR, respectively. Nuclear factor-κB (NF-κB) protein expression was analyzed by western blotting. IL-1β and IL-6 levels were determined by enzyme-linked immunosorbent assay. Blood-brain barrier permeability was determined by Evans blue leakage method. The results showed that baicalin reduced brain edema following ICH in a dose-dependent manner, with concomitant inhibition of NF-κB activation and suppression of MMP-9 expression. In addition, baicalin also reduced IL-1β and IL-6 production, as well as blood-brain barrier permeability. The above results indicated that baicalin prevents against perihematomal edema development after intracerebral hemorrhage possibly through an anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Qing-Bo Zhou
- Department of Neurology, The Second Hospital, Shandong University, Jinan, 250033, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Wang L, Wang F, Wu G, Shi J. Early-stage minimally invasive procedures decrease perihematomal endothelin-1 levels and improve neurological functioning in a rabbit model of intracerebral hemorrhage. Neurol Res 2014; 37:320-7. [PMID: 25258111 DOI: 10.1179/1743132814y.0000000446] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
INTRODUCTION To determine the effects of minimally invasive surgery (MIS) at various stages after intracerebral hemorrhage (ICH) on perihematomal endothelin (ET)-1 levels and neurological functioning. METHODS Sixty rabbits were randomly distributed into a model control group (MC group, 30 rabbits) or a MIS group (MI group, 30 rabbits). An ICH model was established in all animals. In the MI group, ICH was evacuated by MIS at 6, 12, 18, 24, and 48 hours (six rabbits at each time point) after the ICH was established. The animals in the MC group underwent the same procedures for ICH evacuation, but with a sham operation without hematoma aspiration. All the animals were sacrificed 7 days after the ICH was established. Neurological deficit scores were determined, and the perihematomal brain tissue was removed to determine the ET-1 levels, blood-brain barrier (BBB) permeability, and brain water content (BWC). RESULTS The neurological deficit scores, perihematomal ET-1 levels, BBB permeability, and BWC all decreased significantly in the MI group compared to the MC group. Performing the MIS for evacuating the ICH at 6 hours resulted in the most remarkable decreases in these indices, followed by a significant difference observed at 12 hours within the MI subgroups. CONCLUSIONS Performing MIS at 6-12 hours after ICH resulted in the most significant decreases in neurological deficit scores, ET-1 levels, BBB permeability, and brain edema. The optimal time window for performing MIS for ICH evacuation might be within 6-12 hours after hemorrhage.
Collapse
|
47
|
Hatakeyama T, Okauchi M, Hua Y, Keep RF, Xi G. Deferoxamine reduces neuronal death and hematoma lysis after intracerebral hemorrhage in aged rats. Transl Stroke Res 2014; 4:546-53. [PMID: 24187595 DOI: 10.1007/s12975-013-0270-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracerebral hemorrhage (ICH) is primarily a disease of the elderly. Deferoxamine (DFX), an iron chelator, reduces long-term neurological deficits and brain atrophy after ICH in aged rats. In the present study, we investigated whether DFX can reduce acute ICH-induced neuronal death and whether it affects the endogenous response to ICH (ferritin upregulation and hematoma resolution) in aged rats. Male Fischer 344 rats (18 months old) had an intracaudate injection of 100 μL autologous whole blood into the right basal ganglia and were treated with DFX (100 mg/kg) or vehicle 2 hours post-ICH and then every 12 hours up to 7 days. Rats were euthanized 1, 3, or 7 days later for neuronal death, ferritin and hematoma size measurements. Plasma ferritin levels and behavioral outcome following ICH were also examined. DFX treatment significantly reduced ICH-induced neuronal death and neurological deficits. DFX also suppressed ferritin upregulation in the ipsilateral basal ganglia after ICH and hematoma lysis (hematoma volume at day 7: 13.2±4.9 vs. 3.8±1.2 mm3 in vehicle-treated group, p<0.01). However, effects of DFX on plasma ferritin levels after ICH did not reach significance. In conclusion, DFX reduces neuronal death and neurological deficits after ICH in aged rats. It also affects the endogenous response to ICH.
Collapse
Affiliation(s)
- Tetsuhiro Hatakeyama
- Department of Neurosurgery, University of Michigan, USA ; Department of Neurological Surgery, Kagawa University, Japan
| | | | | | | | | |
Collapse
|
48
|
Chen T, Wang J, Li C, Zhang W, Zhang L, An L, Pang T, Shi X, Liao H. Nafamostat mesilate attenuates neuronal damage in a rat model of transient focal cerebral ischemia through thrombin inhibition. Sci Rep 2014; 4:5531. [PMID: 24985053 PMCID: PMC4078306 DOI: 10.1038/srep05531] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/12/2014] [Indexed: 01/01/2023] Open
Abstract
Evidence suggests that thrombin, a blood coagulation serine protease, mediates neuronal injury in experimental cerebral ischemia. Here, we test the hypothesis that nafamostat mesilate, a serine protease inhibitor, may ameliorate ischemia-induced neuronal damage through thrombin inhibition after ischemic stroke. Focal ischemia was induced in adult Sprague-Dawley rats by occlusion of the middle cerebral artery for 2 hours followed by 22 hours of reperfusion. The administration of nafamostat mesilate during ischemia and reperfusion reduced the brain infarct volume, edema volume and neurological deficit. Thrombin expression and activity in the ipsilateral striatum were increased after ischemia, whereas the administration of nafamostat mesilate significantly inhibited thrombin expression and activity. Immunostaining showed that the majority of thrombin was expressed in neurons. TUNEL staining showed that nafamostat mesilate reduced the number of dying cells during ischemia. A rat behavioral test showed that nafamostat mesilate treatment significantly improved the learning ability of ischemic rats. These results suggest that nafamostat mesilate may have a potential therapeutic role for neuroprotection against focal cerebral ischemia through thrombin inhibition.
Collapse
Affiliation(s)
- Tao Chen
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, P.R.China
| | - Jing Wang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, P.R.China
| | - Chenhui Li
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, P.R.China
| | - Weining Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, P.R.China
| | - Luyong Zhang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, P.R.China
| | - Lufan An
- Jiangsu D&R Pharmaceutical Co. LTD, Taizhou 225300, P.R.China
| | - Tao Pang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, P.R.China
| | - Xinzhong Shi
- School of Science, China Pharmaceutical University, Nanjing 210009, P.R.China
| | - Hong Liao
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, P.R.China
| |
Collapse
|
49
|
Zhao J, Chen Z, Xi G, Keep RF, Hua Y. Deferoxamine attenuates acute hydrocephalus after traumatic brain injury in rats. Transl Stroke Res 2014; 5:586-94. [PMID: 24935175 DOI: 10.1007/s12975-014-0353-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/05/2014] [Accepted: 06/05/2014] [Indexed: 11/26/2022]
Abstract
Acute post-traumatic ventricular dilation and hydrocephalus are relatively frequent consequences of traumatic brain injury (TBI). Several recent studies have indicated that high iron levels in brain may relate to hydrocephalus development after intracranial hemorrhage. However, the role of iron in the development of post-traumatic hydrocephalus is still unclear. This study was to determine whether or not iron has a role in hydrocephalus development after TBI. TBI was induced by lateral fluid-percussion in male Sprague-Dawley rats. Some rats had intraventricular injection of iron. Acute hydrocephalus was measured by magnetic resonance T2-weighted imaging and brain hemorrhage was determined by T2* gradient-echo sequence imaging and brain hemoglobin levels. The effect of deferoxamine on TBI-induced hydrocephalus was examined. TBI resulted in acute hydrocephalus at 24 h (lateral ventricle volume: 24.1 ± 3.0 vs. 9.9 ± 0.2 mm(3) in sham group). Intraventricular injection of iron also caused hydrocephalus (25.7 ± 3.4 vs. 9.0 ± 0.6 mm(3) in saline group). Deferoxamine treatment attenuated TBI-induced hydrocephalus and heme oxygenase-1 upregulation. In conclusion, iron may contribute to acute hydrocephalus after TBI.
Collapse
Affiliation(s)
- Jinbing Zhao
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | | | | | | | | |
Collapse
|
50
|
Wu G, Shi J, Wang F, Wang L, Feng A, Ren S. Effects of minimally invasive procedures for evacuation of intracerebral hematoma in early stages on MMP-9 and BBB permeability in rabbits. BMC Neurol 2014; 14:85. [PMID: 24739149 PMCID: PMC4012277 DOI: 10.1186/1471-2377-14-85] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 04/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The effects of performing a minimally invasive procedure at different stages after intracerebral hemorrhage on perihematomal MMP-9 expression and blood-brain barrier (BBB) permeability were evaluated. METHODS Sixty rabbits were randomly distributed into a model control group (MC group, 30 rabbits) or a minimally invasive group (MI group, 30 rabbits). A model of intracerebral hemorrhage was established in the MC and MI group. In the MI group, the intracerebral hematoma was evacuated by stereotactic minimally invasive procedures over 6 hours (6 rabbits), 12 hours (6 rabbits), 18 hours (6 rabbits) 24 hours or 48 hours (6 rabbits) following successful induction of intracerebral hemorrhage. The same procedure was performed in the MC group at the same time point but without evacuating the hematoma. All the animals were sacrificed within two weeks after the hematoma was surgically evacuated. A neurological deficit score was determined, and the perihematomal MMP-9 level and the BBB permeability were measured. RESULTS The neurological deficit score, perihematomal MMP-9 level and BBB permeability of the MI group decreased significantly compared to the MC group. Performing the MI procedure 6-12 h after intracerebral hemorrhage showed the most favorable outcome. CONCLUSIONS Regarding the pathophysiological changes surrounding the hematoma, the optimal time window of performing MI procedures for the intracerebral hematoma evacuation might be within 6-12 h after hemorrhage.
Collapse
Affiliation(s)
- Guofeng Wu
- Emergency Department of Affiliated Hospital, Guiyang Medical College, No, 28, Guiyijie Road, Liuguangmen, Guiyang City, Guizhou Province, 550004 PR China.
| | | | | | | | | | | |
Collapse
|