801
|
Zou X, Wu Z, Zhu W, Chen L, Mao Y, Zhao F. Effectiveness of minocycline in acute white matter injury after intracerebral hemorrhage. J Neurosurg 2016; 126:1855-1862. [PMID: 27494818 DOI: 10.3171/2016.5.jns152670] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Intracerebral hemorrhage (ICH) is a fatal disease with high morbidity and mortality, which may be followed by white matter injury (WMI) due to the local oxidizing reaction induced by iron (Fe). In this study, the authors examined the effect of the tetracycline antibiotic minocycline on Fe-induced WMI and c-Jun N-terminal kinase (JNK) activation in rats. METHODS Thirty-six male Sprague-Dawley rats underwent an intracaudate injection of saline, Fe, or Fe + minocycline. Another 36 rats had an intracaudate injection of autologous blood and were treated with minocycline or vehicle (saline). Biomarkers of both WMI and JNK activation were examined. RESULTS In the Fe-injection group, minocycline suppressed WMI labeled by β-amyloid precursor protein (β-APP) and degraded myelin basic protein (dMBP)/MBP ratio. Protein levels of phosphorylated-JNK were increased after Fe injection, and could be suppressed by minocycline treatment. In the autologous blood-injection group, β-APP and dMBP/MBP levels increased in the ipsilateral site compared with the contralateral site, which could be suppressed by 7 days of minocycline intervention. CONCLUSIONS Iron plays a critical role in WMI after ICH, which can be suppressed by minocycline through reducing the damage induced by Fe.
Collapse
Affiliation(s)
- Xiang Zou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zehan Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fan Zhao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
802
|
Ni H, Shen J, Song Y, Cao M, Liu X, Huang J, Zhang W, Xie L, Ning X, Ke K. EP3, Prostaglandin E2 Receptor Subtype 3, Associated with Neuronal Apoptosis Following Intracerebral Hemorrhage. Cell Mol Neurobiol 2016; 36:971-980. [PMID: 26718710 PMCID: PMC11482414 DOI: 10.1007/s10571-015-0287-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022]
Abstract
EP3 is prostaglandin E2 receptor subtype 3 and mediates the activation of several signaling pathways, changing in cAMP levels, calcium mobilization, and activation of phospholipase C. Previous studies demonstrated a direct role for EP3 in various neurodegenerative disorders, such as stroke and Alzheimer disease. However, the distribution and function of EP3 in ICH diseases remain unknown. Here, we demonstrate that EP3 may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). From the results of Western blot and immunohistochemistry, we obtained a significant up-regulation of EP3 in neurons adjacent to the hematoma following ICH. Up-regulation of EP3 was found to be accompanied by the increased expression of active caspase-3 and pro-apoptotic Bcl-2-associated X protein (Bax) and decreased expression of anti-apoptotic protein B cell lymphoma-2 (Bcl-2) in vivo and vitro studies. Furthermore, the expression of these three proteins reduced active caspase-3 and Bax expression, while increased Bcl-2 were changed after knocking down EP3 by RNA interference in PC12 cells, further confirmed that EP3 might exert its pro-apoptotic function on neuronal apoptosis. Thus, EP3 may play a role in promoting the neuronal apoptosis following ICH.
Collapse
Affiliation(s)
- Haidan Ni
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yan Song
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaorong Liu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jie Huang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Weidong Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Lili Xie
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaojin Ning
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
803
|
Xu Z, Chen J, Shi J, Zhao J, Wang J, Ji Y, Han L, Zhu L, Li X, Zhang D. Upregulated Expression of Karyopherin α2 is Involved in Neuronal Apoptosis Following Intracerebral Hemorrhage in Adult Rats. Cell Mol Neurobiol 2016; 36:755-65. [PMID: 26340948 PMCID: PMC11482370 DOI: 10.1007/s10571-015-0258-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022]
Abstract
Karyopherin α2 (KPNA2) plays a central role in nucleocytoplasmic transport. It is involved in controlling the flow of genetic information and the modulation of diverse cellular activities. Here we explored the KPNA2's roles during the pathophysiological processes of intracerebral hemorrhage (ICH). An ICH rat model was built and evaluated according to behavioral testing. Using Western blot, immunohistochemistry, and immunofluorescence, significant upregulation of KPNA2 was found in neurons in brain areas surrounding the hematoma following ICH. Increasing KPNA2 level was found to be accompanied by the upregulation of active caspase-3, Bax, and decreased expression of Bcl-2. Besides, KPNA2 co-localized well with active caspase-3 in neurons, indicating its potential role in neuronal apoptosis. What's more, knocking down KPNA2 by RNA-interference in PC12 cells reduced active caspase-3 expression. Thus, KPNA2 may play a role in promoting the brain secondary damage following ICH.
Collapse
Affiliation(s)
- Zhiwei Xu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jianping Chen
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jiansheng Shi
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jianmei Zhao
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jun Wang
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuhong Ji
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Lijian Han
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Liang Zhu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaohong Li
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Dongmei Zhang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
804
|
Hu X, Bai X, Zai N, Sun X, Zhu L, Li X. Prognostic value of perfusion-weighted magnetic resonance imaging in acute intracerebral hemorrhage. Neurol Res 2016; 38:614-9. [PMID: 27197990 DOI: 10.1080/01616412.2016.1177932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE This study intends to investigate the prognostic value of perfusion-weighted magnetic resonance imaging in acute intracerebral hemorrhage. METHODS Demographic, clinical and biochemical data between acute intracerebral hemorrhage (AICH) and healthy volunteer groups were assessed in this study, such as rCBV and MTT values. The optimal cutoff values of rCBV and MTT for diagnosing AICH were determined by the ROC curves. Apart from that, we also investigated the association between rCBV/MTT values and cerebral hematoma volumes of AICH patients. The unconditional logistic regression was conducted to determine significant risk factors for AICH. RESULT AICH patients have significantly lower rCBV and higher MTT compared to the control group (all P < 0.05). As suggested by the relatively high sensitivity and specificity, both rCBV and MTT values could be utilized for AICH diagnosis. Moreover, rCBV and MTT were significantly associated with the cerebral hematoma volumes of AICH patients (all P < 0.05). Results from unconditional logistic regression analysis revealed that MTT was a significant risk factor for AICH (P < 0.05 and OR > 1), while rCBV is considered as a protective factor (P < 0.05 and OR < 1). CONCLUSION Perfusion-weighted magnetic resonance imaging produces a high prognostic value for diagnosing AICH.
Collapse
Affiliation(s)
- Xibin Hu
- a Department of Radiology , Affiliated Hospital of Jining Medical University , Jining , China
| | - Xueqin Bai
- a Department of Radiology , Affiliated Hospital of Jining Medical University , Jining , China
| | - Ning Zai
- a Department of Radiology , Affiliated Hospital of Jining Medical University , Jining , China
| | - Xinhai Sun
- a Department of Radiology , Affiliated Hospital of Jining Medical University , Jining , China
| | - Laimin Zhu
- a Department of Radiology , Affiliated Hospital of Jining Medical University , Jining , China
| | - Xian Li
- b Department of Medical Imaging , Jining Medical University , Jining , China
| |
Collapse
|
805
|
Intraventricular Hemorrhage: the Role of Blood Components in Secondary Injury and Hydrocephalus. Transl Stroke Res 2016; 7:447-451. [DOI: 10.1007/s12975-016-0480-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023]
|
806
|
Kim H, Edwards NJ, Choi HA, Chang TR, Jo KW, Lee K. Treatment Strategies to Attenuate Perihematomal Edema in Patients With Intracerebral Hemorrhage. World Neurosurg 2016; 94:32-41. [PMID: 27373415 DOI: 10.1016/j.wneu.2016.06.093] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 11/24/2022]
Abstract
Spontaneous intracerebral hemorrhage (SICH) continues to be a significant cause of neurologic morbidity and mortality throughout the world. Although recent advances in the treatment of SICH have significantly decreased mortality rates, functional recovery has not been dramatically improved by any intervention to date. There are 2 predominant mechanisms of brain injury from intracerebral hemorrhage: mechanical injury from the primary hematoma (including growth of that hematoma), and secondary injury from perihematomal inflammation. For instance, in the hours to weeks after SICH as the hematoma is being degraded, thrombin and iron are released and can result in neurotoxicity, free radical damage, dysregulated coagulation, and harmful inflammatory cascades; this can clinically and radiologically manifest as perihematomal edema (PHE). PHE can contribute to mass effect, cause acute neurologic deterioration in patients, and has even been associated with poor long-term functional outcomes. PHE therefore lends itself to being a potential therapeutic target. In this article, we will review 1) the pathogenesis and time course of the development of PHE, and 2) the clinical series and trials exploring various methods, with a focus on minimally invasive surgical techniques, to reduce PHE and minimize secondary brain injury. Promising areas of continued research also will be discussed.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Neurosurgery, College of Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Nancy J Edwards
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Huimahn A Choi
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Tiffany R Chang
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Kwang Wook Jo
- Department of Neurosurgery, College of Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Kiwon Lee
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
807
|
Sun N, Keep RF, Hua Y, Xi G. Critical Role of the Sphingolipid Pathway in Stroke: a Review of Current Utility and Potential Therapeutic Targets. Transl Stroke Res 2016; 7:420-38. [PMID: 27339463 DOI: 10.1007/s12975-016-0477-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 12/16/2022]
Abstract
Sphingolipids are a series of cell membrane-derived lipids which act as signaling molecules and play a critical role in cell death and survival, proliferation, recognition, and migration. Sphingosine-1-phosphate acts as a key signaling molecule and regulates lymphocyte trafficking, glial cell activation, vasoconstriction, endothelial barrier function, and neuronal death pathways which plays a critical role in numerous neurological conditions. Stroke is a second leading cause of death all over the world and effective therapies are still in great demand, including ischemic stroke and hemorrhagic stroke as well as poststroke repair. Significantly, sphingolipid activities change after stroke and correlate with stroke outcome, which has promoted efforts to testify whether the sphingolipid pathway could be a novel therapeutic target in stroke. The sphingolipid metabolic pathway, the connection between the pathway and stroke, as well as therapeutic interventions to manipulate the pathway to reduce stroke-induced brain injury are discussed in this review.
Collapse
Affiliation(s)
- Na Sun
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
808
|
Han X, Lan X, Li Q, Gao Y, Zhu W, Cheng T, Maruyama T, Wang J. Inhibition of prostaglandin E2 receptor EP3 mitigates thrombin-induced brain injury. J Cereb Blood Flow Metab 2016; 36:1059-74. [PMID: 26661165 PMCID: PMC4908617 DOI: 10.1177/0271678x15606462] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 07/27/2015] [Indexed: 11/16/2022]
Abstract
Prostaglandin E2 EP3 receptor is the only prostaglandin E2 receptor that couples to multiple G-proteins, but its role in thrombin-induced brain injury is unclear. In the present study, we exposed mouse hippocampal slice cultures to thrombin in vitro and injected mice with intrastriatal thrombin in vivo to investigate the role of EP3 receptor in thrombin-induced brain injury and explore its underlying cellular and molecular mechanisms. In vitro, EP3 receptor inhibition reduced thrombin-induced hippocampal CA1 cell death. In vivo, EP3 receptor was expressed in astrocytes and microglia in the perilesional region. EP3 receptor inhibition reduced lesion volume, neurologic deficit, cell death, matrix metalloproteinase-9 activity, neutrophil infiltration, and the number of CD68(+) microglia, but increased the number of Ym-1(+) M2 microglia. RhoA-Rho kinase levels were increased after thrombin injection and were decreased by EP3 receptor inhibition. In mice that received an intrastriatal injection of autologous arterial blood, inhibition of thrombin activity with hirudin decreased RhoA expression compared with that in vehicle-treated mice. However, EP3 receptor activation reversed this effect of hirudin. These findings show that prostaglandin E2 EP3 receptor contributes to thrombin-induced brain damage via Rho-Rho kinase-mediated cytotoxicity and proinflammatory responses.
Collapse
Affiliation(s)
- Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiang Li
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yufeng Gao
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wei Zhu
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tian Cheng
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takayuki Maruyama
- Project Management, Discovery and Research, Ono Pharmaceutical Co. Ltd., Mishima-gun, Osaka, Japan
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
809
|
Abstract
About half of patients survive intracerebral hemorrhage (ICH), but most are left with significant disability. Rehabilitation after ICH is the mainstay of treatment to reduce impairment, improve independence in activities, and return patients to meaningful participation in the community. The authors discuss the neuroplastic mechanisms underlying recovery in ICH, preclinical and clinical interventional studies to augment recovery, and the rehabilitative and medical management of post-ICH patients.
Collapse
Affiliation(s)
- Michael F Saulle
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York
| | - Heidi M Schambra
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York
| |
Collapse
|
810
|
Askenase MH, Sansing LH. Stages of the Inflammatory Response in Pathology and Tissue Repair after Intracerebral Hemorrhage. Semin Neurol 2016; 36:288-97. [PMID: 27214704 DOI: 10.1055/s-0036-1582132] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major health concern, with high rates of mortality and morbidity and no highly effective clinical interventions. Basic research in animal models of ICH has provided insight into its complex pathology, in particular revealing the role of inflammation in driving neuronal death and neurologic deficits after hemorrhage. The response to ICH occurs in four distinct phases: (1) initial tissue damage and local activation of inflammatory factors, (2) inflammation-driven breakdown of the blood-brain barrier, (3) recruitment of circulating inflammatory cells and subsequent secondary immunopathology, and (4) engagement of tissue repair responses that promote tissue repair and restoration of neurologic function. The development of CNS inflammation occurs over many days after initial hemorrhage and thus may represent an ideal target for treatment of the disease, but further research is required to identify the mechanisms that promote engagement of inflammatory versus anti-inflammatory pathways. In this review, the authors examine how experimental models of ICH have uncovered critical mediators of pathology in each of the four stages of the inflammatory response, and focus on the role of the immune system in these processes.
Collapse
Affiliation(s)
- Michael H Askenase
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
811
|
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
|
812
|
Tempol alleviates intracerebral hemorrhage-induced brain injury possibly by attenuating nitrative stress. Neuroreport 2016; 26:842-9. [PMID: 26237245 DOI: 10.1097/wnr.0000000000000434] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Intracerebral hemorrhage (ICH)-induced brain injury leads to irreversible disruption of the blood-brain barrier (BBB) and fatality brain edema with massive cell death. Although secondary damage could, in principle, be preventable, no effective treatment approaches currently exist for patients with ICH. Tempol, a catalytic scavenger of peroxynitrite (ONOO)-derived free radicals, has been proven to ameliorate brain injury in several types of brain insults. This study aims to investigate the potential neuroprotective effect of tempol after ICH and to explore the underlying mechanisms. Collagenase-induced ICH was performed in rats. Tempol was administered immediately after ICH. The effects of tempol on ICH were evaluated by assessing neurological deficits, BBB permeability, brain edema, and apoptotic cell death. The mechanisms of action of tempol, with its clear ability on the derivative of ONOO [3-nitrotyrosine (3-NT), ONOO, and its derivative-mediated nitration marker] and expression of tight junction protein [zonula occludens-1 (ZO-1)], were also investigated. Perihematomal 3-NT increased significantly following ICH and expressed around vessels accompanied by reduced and discontinuous expression of ZO-1. Tempol treatment significantly suppressed 3-NT formation and preserved ZO-1 levels, and led to improvement in neurological outcomes and reduction of BBB leakiness, brain edema, and apoptosis. In conclusion, tempol has neuroprotective potential in experimental ICH and may help combat ICH-induced brain injury in patients.
Collapse
|
813
|
Sun N, Shen Y, Han W, Shi K, Wood K, Fu Y, Hao J, Liu Q, Sheth KN, Huang D, Shi FD. Selective Sphingosine-1-Phosphate Receptor 1 Modulation Attenuates Experimental Intracerebral Hemorrhage. Stroke 2016; 47:1899-906. [PMID: 27174529 DOI: 10.1161/strokeaha.115.012236] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Preclinical studies and a proof-of-concept clinical study have shown that sphingosine-1-phosphate receptor (S1PR) modulator, fingolimod, improves the clinical outcome of intracerebral hemorrhage (ICH). However, the specific subtype of the S1PRs through which immune modulation provides protection in ICH remains unclear. In addition, fingolimod-induced adverse effects could limit its use in patients with stroke because of interactions with other S1PR subtypes, particularly with S1PR3. RP101075 is a selective S1PR1 agonist with superior cardiovascular safety profile. In this study, we investigated the impact of RP101075 treatment in a mouse model of ICH. METHODS ICH was induced by injection of autologous blood in 294 male C57BL/6J and Rag2(-/-) mice. ICH mice randomly received vehicle, RP101075, or RP101075 plus S1PR1 antagonist W146 by daily oral gavage for three consecutive days, starting from 30 minutes after surgery. Neurodeficits, brain edema, brain infiltration of immune cells, blood-brain barrier integrity, and cell death were assessed after ICH. RESULTS RP101075 significantly attenuated neurological deficits and reduced brain edema in ICH mice. W146 blocked the effects of RP101075 on neurodeficits and brain edema. RP101075 reduced the counts of brain-infiltrating lymphocytes, neutrophils, and microglia, as well as cytokine expression after ICH. Enhanced blood-brain barrier integrity and alleviated neuronal death were also seen in ICH mice after RP101075 treatment. CONCLUSIONS S1PR1 modulation via RP101075 provides protection in experimental ICH. Together with the advantageous pharmacological features of RP101075, these results warrant further investigations of its mechanisms of action and translational values in ICH patients.
Collapse
Affiliation(s)
- Na Sun
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Yi Shen
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Wei Han
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Kaibin Shi
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Kristofer Wood
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Ying Fu
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Junwei Hao
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Qiang Liu
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Kevin N Sheth
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - DeRen Huang
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Fu-Dong Shi
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.).
| |
Collapse
|
814
|
The Injury and Therapy of Reactive Oxygen Species in Intracerebral Hemorrhage Looking at Mitochondria. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2592935. [PMID: 27293511 PMCID: PMC4880716 DOI: 10.1155/2016/2592935] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/20/2016] [Accepted: 04/17/2016] [Indexed: 01/12/2023]
Abstract
Intracerebral hemorrhage is an emerging major health problem often resulting in death or disability. Reactive oxygen species (ROS) have been identified as one of the major damaging factors in ischemic stroke. However, there is less discussion about ROS in hemorrhage stroke. Metabolic products of hemoglobin, excitatory amino acids, and inflammatory cells are all sources of ROS, and ROS harm the central nervous system through cell death and structural damage, especially disruption of the blood-brain barrier. We have considered the antioxidant system of the CNS itself and the drugs aiming to decrease ROS after ICH, and we find that mitochondria are key players in all of these aspects. Moreover, when the mitochondrial permeability transition pore opens, ROS-induced ROS release, which leads to extensive liberation of ROS and mitochondrial failure, occurs. Therefore, the mitochondrion may be a significant target for elucidating the problem of ROS in ICH; however, additional experimental support is required.
Collapse
|
815
|
Cao S, Zheng M, Hua Y, Chen G, Keep RF, Xi G. Hematoma Changes During Clot Resolution After Experimental Intracerebral Hemorrhage. Stroke 2016; 47:1626-31. [PMID: 27125525 DOI: 10.1161/strokeaha.116.013146] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/29/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND PURPOSE Hematoma clearance occurs in the days after intracerebral hemorrhage (ICH) and has not been well studied. In the current study, we examined changes in the hematoma in a piglet ICH model. The effect of deferoxamine on hematoma was also examined. METHODS The ICH model was induced by an injection of autologous blood into the right frontal lobe of piglets. First, a natural time course of hematoma changes ≤7 days was determined. Second, the effect of deferoxamine on hematoma changes was examined. Hemoglobin and membrane attack complex levels in the hematoma were examined by enzyme-linked immunosorbent assay. Immunohistochemistry and Western blotting were used to examine CD47 (a regulator of erythrophagocytosis), CD163 (a hemoglobin scavenger receptor), and heme oxygenase-1 (a heme degradation enzyme) in the clot. RESULTS After ICH, there was a reduction in red blood cell diameter within the clot with time. This was accompanied by membrane attack complex accumulation and decreased hemoglobin levels. Erythrophagocytosis occurred in the hematoma, and this was associated with reduced clot CD47 levels. Activated macrophages/microglia were CD163 and hemeoxygenase-1 positive, and these accumulated in the clot with time. Deferoxamine treatment attenuated the process of hematoma resolution by reducing member attack complex formation and inhibiting CD47 loss in the clot. CONCLUSIONS These results indicate that membrane attack complex and erythrophagocytosis contribute to hematoma clearance after ICH, which can be altered by deferoxamine treatment.
Collapse
Affiliation(s)
- Shenglong Cao
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (S.C., M.Z., Y.H., R.F.K., G.X.); and Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C., G.C.)
| | - Mingzhe Zheng
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (S.C., M.Z., Y.H., R.F.K., G.X.); and Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C., G.C.)
| | - Ya Hua
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (S.C., M.Z., Y.H., R.F.K., G.X.); and Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C., G.C.)
| | - Gao Chen
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (S.C., M.Z., Y.H., R.F.K., G.X.); and Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C., G.C.)
| | - Richard F Keep
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (S.C., M.Z., Y.H., R.F.K., G.X.); and Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C., G.C.)
| | - Guohua Xi
- From the Department of Neurosurgery, University of Michigan, Ann Arbor (S.C., M.Z., Y.H., R.F.K., G.X.); and Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C., G.C.).
| |
Collapse
|
816
|
Hu L, Dong MX, Zhao H, Xu GH, Qin XY. Fibulin-5: a novel biomarker for evaluating severity and predicting prognosis in patients with acute intracerebral haemorrhage. Eur J Neurol 2016; 23:1195-201. [PMID: 27106135 DOI: 10.1111/ene.13013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022]
Affiliation(s)
- L. Hu
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
- Department of Neurology; the Fifth People's Hospital of Chongqing; Chongqing China
| | - M.-X. Dong
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - H. Zhao
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - G.-H. Xu
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - X.-Y. Qin
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| |
Collapse
|
817
|
Morotti A, Phuah CL, Anderson CD, Jessel MJ, Schwab K, Ayres AM, Pezzini A, Padovani A, Gurol ME, Viswanathan A, Greenberg SM, Goldstein JN, Rosand J. Leukocyte Count and Intracerebral Hemorrhage Expansion. Stroke 2016; 47:1473-8. [PMID: 27103016 DOI: 10.1161/strokeaha.116.013176] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/22/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE Acute leukocytosis is a well-established response to intracerebral hemorrhage (ICH). Leukocytes, because of their interaction with platelets and coagulation factors, may in turn play a role in hemostasis. We investigated whether admission leukocytosis was associated with reduced bleeding after acute ICH. METHODS Consecutive patients with primary ICH were prospectively collected from 1994 to 2015 and retrospectively analyzed. We included subjects with a follow-up computed tomographic scan available and automated complete white blood cell count performed within 48 hours from onset. Baseline and follow-up hematoma volumes were calculated with semiautomated software, and hematoma expansion was defined as volume increase >30% or 6 mL. The association between white blood cell count and ICH expansion was investigated with multivariate logistic regression. RESULTS A total of 1302 subjects met eligibility criteria (median age, 75 years; 55.8% men), of whom 207 (15.9%) experienced hematoma expansion. Higher leukocyte count on admission was associated with reduced risk of hematoma expansion (odds ratio for 1000 cells increase, 0.91; 95% confidence interval, 0.86-0.96; P=0.001). The risk of hematoma expansion was inversely associated with neutrophil count (odds ratio, 0.90; 95% confidence interval, 0.85-0.96; P=0.001) and directly associated with monocyte count (odds ratio, 2.71; 95% confidence interval, 1.08-6.83; P=0.034). There was no association between lymphocyte count and ICH expansion (odds ratio, 0.96; 95% confidence interval, 0.79-1.17; P=0.718). CONCLUSIONS Higher admission white blood cell count is associated with lower risk of hematoma expansion. This highlights a potential role of the inflammatory response in modulating the coagulation cascade after acute ICH.
Collapse
Affiliation(s)
- Andrea Morotti
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston.
| | - Chia-Ling Phuah
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Christopher D Anderson
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Michael J Jessel
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Kristin Schwab
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Alison M Ayres
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Alessandro Pezzini
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Alessandro Padovani
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - M Edip Gurol
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Anand Viswanathan
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Steven M Greenberg
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Joshua N Goldstein
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| | - Jonathan Rosand
- From the Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy (A.M., A. Pezzini, A. Padovani); Division of Neurocritical Care and Emergency Neurology, Department of Neurology (A.M., C.-L.P., C.D.A., M.J.J., J.N.G., J.R.), Hemorrhagic Stroke Research Center (A.M., C.-L.P., C.D.A., M.J.J., K.S., A.A., M.E.G., A.V., S.M.G., J.N.G., J.R.), and Department of Emergency Medicine (J.N.G.), Massachusetts General Hospital, Boston
| |
Collapse
|
818
|
Wu G, Wu J, Wang L, Jiao Y, Zhou H, Tang Z. Minimally invasive surgery for ICH evacuation followed by rosiglitazone infusion therapy increased perihematomal PPARγ expression and improved neurological outcomes in rabbits. Neurol Res 2016; 38:261-8. [PMID: 27082035 DOI: 10.1080/01616412.2015.1105627] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To observe the effects of minimally invasive surgery (MIS) for intracerebral hematoma (ICH) evacuation followed by rosiglitazone infusion therapy on peroxisome proliferator-activated receptor-gamma (PPARγ), blood-brain barrier (BBB) permeability, and neurological function. METHODS A total of 75 male rabbits (2.8-3.4 kg) were randomly assigned to a normal control group (NC group), a model control group (MC group), a rosiglitazone group (RSG group), a minimally invasive treatment group (MIS group) or a MIS combined with rosiglitazone group (MIS+RSG group). ICH was induced in all of the animals except for those in the NC group. The rosiglitazone was infused into the hematoma area in the RSG group and the MIS+RSG group. A MIS was performed to evacuate the ICH 6 h after the successful preparation of the ICH model in the MIS group and the MIS+RSG group. Each group included 15 rabbits and was divided equally into 3 subgroups (each subgroup included 5 rabbits that were killed on day 1, day 3, or day 7). Neurological deficit scores were determined, and the perihematomal brain tissue was removed to determine the PPARγ level and BBB permeability. RESULTS Neurological deficit scores, perihematomal PPARγ levels, and BBB permeability were all significantly increased in the MC group compared to the NC group. Performing the MIS alone to evacuate the ICH resulted in a marked decrease in these indices. The RSG used alone increased PPARγ levels and decreased BBB disruption. The MIS+RSG group displayed a marked increase in PPARγ levels and a more significant decrease in BBB permeability and neurological deficit scores. CONCLUSIONS Performing MIS followed by PPARγ agonist infusion therapy is more efficacious for reducing secondary damage to the brain and improving neurological function.
Collapse
Affiliation(s)
- Guofeng Wu
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Junjie Wu
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Likun Wang
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Yu Jiao
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Houguang Zhou
- b Department of Neurology , Affiliated Huashan Hospital of Fudan University , Shanghai , China
| | - Zhouping Tang
- c Department of Neurology , Affiliated Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan City , Hubei Province , China
| |
Collapse
|
819
|
Intracerebral Hemorrhage, Oxidative Stress, and Antioxidant Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1203285. [PMID: 27190572 PMCID: PMC4848452 DOI: 10.1155/2016/1203285] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/20/2015] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
Hemorrhagic stroke is a common and severe neurological disorder and is associated with high rates of mortality and morbidity, especially for intracerebral hemorrhage (ICH). Increasing evidence demonstrates that oxidative stress responses participate in the pathophysiological processes of secondary brain injury (SBI) following ICH. The mechanisms involved in interoperable systems include endoplasmic reticulum (ER) stress, neuronal apoptosis and necrosis, inflammation, and autophagy. In this review, we summarized some promising advances in the field of oxidative stress and ICH, including contained animal and human investigations. We also discussed the role of oxidative stress, systemic oxidative stress responses, and some research of potential therapeutic options aimed at reducing oxidative stress to protect the neuronal function after ICH, focusing on the challenges of translation between preclinical and clinical studies, and potential post-ICH antioxidative therapeutic approaches.
Collapse
|
820
|
Zhang RZ, Tao CY, Chen W, Wang CH, Hu Y, Song L, Zhang B, Chen YS, Xu ZQ, Wang L, Feng H, Wang TH, Zheng J, You C, Gao FB. Dynamic Diffusion Tensor Imaging Reveals Structural Changes in the Bilateral Pyramidal Tracts after Brain Stem Hemorrhage in Rats. Front Neuroanat 2016; 10:33. [PMID: 27065816 PMCID: PMC4811861 DOI: 10.3389/fnana.2016.00033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 03/14/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Few studies have concentrated on pyramidal tract (PY) changes after brain stem hemorrhage (BSH). In this study, we used a diffusion tensor imaging (DTI) technique and histologic identification to investigate longitudinal PY changes on both the contralateral and ipsilateral sides after experimental BSH. METHODS BSH was induced in 61 Sprague-Dawley rats by infusing 30 μl of autogenous tail blood into each rat's right pons. DTI and motor function examinations were performed repeatedly on days 1, 3, 7, 14, and 28 after surgery. Fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity, and radial diffusivity were measured in the bilateral PYs. The axon and myelin injury in the PY were evaluated by histologic study. RESULTS As compared with normal controls, the bilateral PYs in rats with induced BSH showed an early decrease and a late increase in FA and an early increase and a late decrease in MD. A progressive decrease in axial diffusivity with dramatic axon loss from day 1 to day 28 after BSH was found bilaterally. The bilateral PYs showed an early increase and a late decrease in radial diffusivity. Early myelin injury and late repair were also detected pathologically in the bilateral PYs of rats with BSH. Thus, the early motor function deficits of rats with BSH began to improve on day 14 and had almost completely disappeared by day 28. CONCLUSIONS DTI revealed dynamic changes in the bilateral PYs after BSH, which was confirmed by histologic findings and which correlated with motor function alteration. These findings support the idea that quantitative DTI can track structural changes in the bilateral PYs and that DTI may serve as a noninvasive tool to predict the prognoses of patients with BSH.
Collapse
Affiliation(s)
- Ru-Zhi Zhang
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Chuan-Yuan Tao
- Department of Neurosurgery, West China Hospital, Sichuan University Chengdu, China
| | - Wei Chen
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Chun-Hua Wang
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Yue Hu
- Department of Anesthesiology and Institute of Neurological Disease, Translation Neuroscience Center, West China Hospital, Sichuan University Chengdu, China
| | - Li Song
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Bing Zhang
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Yu-Shu Chen
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Zi-Qian Xu
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Lei Wang
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University Chongqing, China
| | - Ting-Hua Wang
- Department of Anesthesiology and Institute of Neurological Disease, Translation Neuroscience Center, West China Hospital, Sichuan University Chengdu, China
| | - Jie Zheng
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO, USA
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University Chengdu, China
| | - Fa-Bao Gao
- Department of Radiology, West China Hospital, Sichuan University Chengdu, China
| |
Collapse
|
821
|
Cui Z, Zhong Z, Yang Y, Wang B, Sun Y, Sun Q, Yang GY, Bian L. Ferrous Iron Induces Nrf2 Expression in Mouse Brain Astrocytes to Prevent Neurotoxicity. J Biochem Mol Toxicol 2016; 30:396-403. [PMID: 27037625 DOI: 10.1002/jbt.21803] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/22/2016] [Accepted: 03/05/2016] [Indexed: 11/09/2022]
Abstract
Free radical damage caused by ferrous iron is involved in the pathogenesis of secondary brain injury after intracerebral hemorrhage (ICH). NF-E2-related factor 2 (Nrf2), a major phase II gene regulator that binds to antioxidant response element, represents an important cellular cytoprotective mechanism against oxidative damage. We hypothesized that Nrf2 might protect astrocytes from damage by Fe(2+) . Therefore, we examined cytotoxicity in primary astrocytes induced by iron overload and evaluated the effects of Fe(2+) on Nrf2 expression. The results demonstrated that 24-h Fe(2+) exposure exerted time- and concentration-dependent cytotoxicity in astrocytes. Furthermore, Fe(2+) exposure in astrocytes resulted in time- and concentration-dependent increases in Nrf2 expression, which preceded Fe(2+) toxicity. Nrf2-specific siRNA further knocked down Nrf2 levels, resulting in greater Fe(2+) -induced astrocyte cytotoxicity. These data indicate that induction of Nrf2 expression could serve as an adaptive self-defense mechanism, although it is insufficient to completely protect primary astrocytes from Fe(2+) -induced neurotoxicity.
Collapse
Affiliation(s)
- Zhenwen Cui
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhihong Zhong
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yong Yang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Baofeng Wang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuhao Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingfang Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Department of Neurosurgery, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
822
|
Weinstock MJ, Uhlmann EJ, Zwicker JI. Intracranial hemorrhage in cancer patients treated with anticoagulation. Thromb Res 2016; 140 Suppl 1:S60-5. [DOI: 10.1016/s0049-3848(16)30100-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
823
|
Salihu AT, Muthuraju S, Idris Z, Izaini Ghani AR, Abdullah JM. Functional outcome after intracerebral haemorrhage – a review of the potential role of antiapoptotic agents. Rev Neurosci 2016; 27:317-27. [DOI: 10.1515/revneuro-2015-0046] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/21/2015] [Indexed: 11/15/2022]
Abstract
AbstractIntracerebral haemorrhage (ICH) is the second most common form of stroke and is associated with greater mortality and morbidity compared with ischaemic stroke. The current ICH management strategies, which mainly target primary injury mechanisms, have not been shown to improve patient’s functional outcome. Consequently, multimodality treatment approaches that will focus on both primary and secondary pathophysiology have been suggested. During the last decade, a proliferation of experimental studies has demonstrated the role of apoptosis in secondary neuronal loss at the periphery of the clot after ICH. Subsequently, the value of certain antiapoptotic agents in reducing neuronal death and improving functional outcome following ICH was evaluated in animal models. Preliminary evidence from those studies strongly supports the potential role of antiapoptotic agents in reducing neuronal death and improving functional outcome after intracerebral haemorrhage. Expectedly, the ongoing and subsequent clinical trials will substantiate these findings and provide clear information on the most potent and safe antiapoptotic agents, their appropriate dosage, and temporal window of action, thereby making them suitable for the multimodality treatment approach.
Collapse
|
824
|
Wu CH, Chen CC, Lai CY, Hung TH, Lin CC, Chao M, Chen SF. Treatment with TO901317, a synthetic liver X receptor agonist, reduces brain damage and attenuates neuroinflammation in experimental intracerebral hemorrhage. J Neuroinflammation 2016; 13:62. [PMID: 26968836 PMCID: PMC4788882 DOI: 10.1186/s12974-016-0524-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/03/2016] [Indexed: 11/10/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) induces a series of inflammatory processes that contribute to neuronal damage and neurological deterioration. Liver X receptors (LXRs) are nuclear receptors that negatively regulate transcriptional processes involved in inflammatory responses, but their role in the pathology following ICH remains unclear. The present study investigated the neuroprotective effects and anti-inflammatory actions of TO901317, a synthetic LXR agonist, in a model of collagenase-induced ICH and in microglial cultures. Methods Mice subjected to collagenase-induced ICH injury were injected with either TO901317 (30 mg/kg) or vehicle 10 min after ICH and subsequently daily for 2 days. Behavioral studies, histology analysis, and assessments of hematoma volumes, brain water content, and blood-brain barrier (BBB) permeability were performed. The protein expression of LXR-α, LXR-β, ATP binding cassette transporter-1 (ABCA-1), and inflammatory molecules was analyzed. The anti-inflammatory mechanism of TO901317 was investigated in cultured microglia that were stimulated with either lipopolysaccharide (LPS) or thrombin. Results ICH induced an increase in LXR-α protein levels in the hemorrhagic hemisphere at 6 h whereas LXR-β expression remained unaffected. Both LXR-α and LXR-β were expressed in neurons and microglia in the peri-ICH region and but rarely in astrocytes. TO901317 significantly attenuated functional deficits and brain damage up to 28 days post-ICH. TO901317 also reduced neuronal death, BBB disruption, and brain edema at day 4 post-ICH. These changes were associated with marked reductions in microglial activation, neutrophil infiltration, and expression levels of inflammatory mediators at 4 and 7 days. However, TO901317 had no effect on matrix metalloproteinase-9 activity. In BV2 microglial cultures, TO901317 attenuated LPS- and thrombin-stimulated nitric oxide production and reduced LPS-induced p38, JNK, MAPK, and nuclear factor-kappa B (NF-κB) signaling. Moreover, delaying administration of TO901317 to 3 h post-ICH reduced brain tissue damage and neuronal death. Conclusions Our results suggest that enhancing LXR activation may provide a potential therapy for ICH by modulating the cytotoxic functions of microglia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0524-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chun-Hu Wu
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chai-You Lai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Taipei and College of Medicine, Chang Gung University, Taipei, Taiwan, Republic of China
| | - Chao-Chang Lin
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Min Chao
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China.
| |
Collapse
|
825
|
Jiang C, Zuo F, Wang Y, Wan J, Yang Z, Lu H, Chen W, Zang W, Yang Q, Wang J. Progesterone exerts neuroprotective effects and improves long-term neurologic outcome after intracerebral hemorrhage in middle-aged mice. Neurobiol Aging 2016; 42:13-24. [PMID: 27143417 DOI: 10.1016/j.neurobiolaging.2016.02.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/27/2016] [Accepted: 02/28/2016] [Indexed: 11/26/2022]
Abstract
In this study, we examined the effect of progesterone on histopathologic and functional outcomes of intracerebral hemorrhage (ICH) in 10- to 12-month-old mice. Progesterone or vehicle was administered by intraperitoneal injection 1 hour after collagenase-induced ICH and then by subcutaneous injections at 6, 24, and 48 hours. Oxidative and nitrosative stress were assayed at 12 hours post-ICH. Injury markers were examined on day 1, and lesion was examined on day 3. Neurologic deficits were examined for 28 days. Progesterone posttreatment reduced lesion volume, brain swelling, edema, and cell degeneration and improved long-term neurologic function. These protective effects were associated with reductions in protein carbonyl formation, protein nitrosylation, and matrix metalloproteinase-9 activity and attenuated cellular and molecular inflammatory responses. Progesterone also reduced vascular endothelial growth factor expression, increased neuronal-specific Na(+)/K(+) ATPase ɑ3 subunit expression, and reduced protein kinase C-dependent Na(+)/K(+) ATPase phosphorylation. Furthermore, progesterone reduced glial scar thickness, myelin loss, brain atrophy, and residual injury volume on day 28 after ICH. With multiple brain targets, progesterone warrants further investigation for its potential use in ICH therapy.
Collapse
Affiliation(s)
- Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China; Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Fangfang Zuo
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Yuejuan Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Jieru Wan
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Zengjin Yang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Wenwu Chen
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Weidong Zang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jian Wang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
826
|
Stokum JA, Gerzanich V, Simard JM. Molecular pathophysiology of cerebral edema. J Cereb Blood Flow Metab 2016; 36:513-38. [PMID: 26661240 PMCID: PMC4776312 DOI: 10.1177/0271678x15617172] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 12/25/2022]
Abstract
Advancements in molecular biology have led to a greater understanding of the individual proteins responsible for generating cerebral edema. In large part, the study of cerebral edema is the study of maladaptive ion transport. Following acute CNS injury, cells of the neurovascular unit, particularly brain endothelial cells and astrocytes, undergo a program of pre- and post-transcriptional changes in the activity of ion channels and transporters. These changes can result in maladaptive ion transport and the generation of abnormal osmotic forces that, ultimately, manifest as cerebral edema. This review discusses past models and current knowledge regarding the molecular and cellular pathophysiology of cerebral edema.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA Department of Pathology, University of Maryland School of Medicine, Baltimore, USA Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
827
|
Abstract
OPINION STATEMENT Cerebral edema (i.e., "brain swelling") is a common complication following intracerebral hemorrhage (ICH) and is associated with worse clinical outcomes. Perihematomal edema (PHE) accumulates during the first 72 h after hemorrhage, and during this period, patients are at risk of clinical deterioration due to the resulting tissue shifts and brain herniation. First-line medical therapies for patients symptomatic of PHE include osmotic agents, such as mannitol in low- or high-dose bolus form, or boluses of hypertonic saline (HTS) at varied concentrations with or without subsequent continuous infusion. Decompressive craniectomy may be required for symptomatic edema refractory to osmotherapy. Other strategies that reduce PHE such as hypothermia and minimally invasive surgery have shown promise in pilot studies and are currently being evaluated in larger clinical trials. Ongoing basic, translational, and clinical research seek to better elucidate the pathophysiology of PHE to identify novel strategies to prevent edema formation as a next major advance in the treatment of ICH.
Collapse
|
828
|
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
|
829
|
Orakcioglu B, Kentar MM, Schiebel P, Uozumi Y, Unterberg A, Sakowitz OW. Perihemorrhagic ischemia occurs in a volume-dependent manner as assessed by multimodal cerebral monitoring in a porcine model of intracerebral hemorrhage. Neurocrit Care 2016; 22:133-9. [PMID: 25052158 DOI: 10.1007/s12028-014-0027-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Changes in the perihemorrhagic zone (PHZ) of intracerebral hemorrhage (ICH) are variable. Different mechanisms contribute to secondary neuronal injury after ICH. This multimodal monitoring study investigated early changes in the PHZ of ICH. METHODS Twenty-four swine were anesthetized, ventilated, and underwent monitoring of vital parameters. Next to an intracranial pressure-probe (ICP), microdialysis (MD), thermodiffusion cerebral blood flow (td-CBF), and oxygen probes (PbrO2) were placed into the gray white matter junction for 12 h of monitoring. ICH was induced using the autologous blood injection model. Pre-defined volumes were 0 ml (sham), 1.5 ml ipsilateral (1.5 ml), 3.0 ml ipsilateral (3.0 ml), and 3.0 ml contralateral (3.0 ml contra). RESULTS ICP equally increased in all groups after ICH. In the 3.0 ml group tissue oxygenation decreased to ischemic values of 9 ± 7 mmHg early after 6 h of monitoring. This decrease was associated with a significant perfusion reduction from 36 ± 8 ml/100 g/min to 20 ± 10 ml/100 g/min. MD correlated with a threefold lactate/pyruvate ratio increase. Measurements in all other groups were unchanged. CONCLUSION Multimodal monitoring demonstrates volume-dependent changes of tissue oxygenation, blood flow, and ischemic MD markers in the PHZ independent of increased ICP suggesting early moderate ischemia. No evidence was found for the existence of a perihemorrhagic ischemia in the small hematoma groups.
Collapse
Affiliation(s)
- Berk Orakcioglu
- Department of Neurosurgery, Ruprecht-Karls-University, Heidelberg, Germany,
| | | | | | | | | | | |
Collapse
|
830
|
After Intracerebral Hemorrhage, Oligodendrocyte Precursors Proliferate and Differentiate Inside White-Matter Tracts in the Rat Striatum. Transl Stroke Res 2016; 7:192-208. [PMID: 26743212 PMCID: PMC4873533 DOI: 10.1007/s12975-015-0445-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/17/2015] [Accepted: 12/28/2015] [Indexed: 02/06/2023]
Abstract
Damage to myelinated axons contributes to neurological deficits after acute CNS injury, including ischemic and hemorrhagic stroke. Potential treatments to promote re-myelination will require fully differentiated oligodendrocytes, but almost nothing is known about their fate following intracerebral hemorrhage (ICH). Using a rat model of ICH in the striatum, we quantified survival, proliferation, and differentiation of oligodendrocyte precursor cells (OPCs) (at 1, 3, 7, 14, and 28 days) in the peri-hematoma region, surrounding striatum, and contralateral striatum. In the peri-hematoma, the density of Olig2+ cells increased dramatically over the first 7 days, and this coincided with disorganization and fragmentation of myelinated axon bundles. Very little proliferation (Ki67+) of Olig2+ cells was seen in the anterior subventricular zone from 1 to 28 days. However, by 3 days, many were proliferating in the peri-hematoma region, suggesting that local proliferation expands their population. By 14 days, the density of Olig2+ cells declined in the peri-hematoma region, and, by 28 days, it reached the low level seen in the contralateral striatum. At these later times, many surviving axons were aligned into white-matter bundles, which appeared less swollen or fragmented. Oligodendrocyte cell maturation was prevalent over the 28-day period. Densities of immature OPCs (NG2+Olig2+) and mature (CC-1+Olig2+) oligodendrocytes in the peri-hematoma increased dramatically over the first week. Regardless of the maturation state, they increased preferentially inside the white-matter bundles. These results provide evidence that endogenous oligodendrocyte precursors proliferate and differentiate in the peri-hematoma region and have the potential to re-myelinate axon tracts after hemorrhagic stroke.
Collapse
|
831
|
Chen JW, Paff MR, Abrams-Alexandru D, Kaloostian SW. Decreasing the Cerebral Edema Associated with Traumatic Intracerebral Hemorrhages: Use of a Minimally Invasive Technique. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:279-284. [PMID: 26463961 DOI: 10.1007/978-3-319-18497-5_48] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Traumatic brain injury (TBI) is a major public health problem worldwide that affects all age groups. In the United States alone, there are approximately 50,000 deaths from severe traumatic brain injuries each year. In most studies, about 40 % of severe TBI have associated traumatic intracerebral hemorrhages (tICHs). The surgical treatment of tICH is debated largely because of its invasive nature, particularly in reaching deep tICHs. tICHs have a clear contribution to mass effect and exacerbate cerebral edema and ICP because of the break-down products of hemorrhage. We introduce a modification of the Mi SPACE technique (Minimally Invasive Subcortical Parafascicular Transsulcal Access for Clot Evacuation) that is applicable to tICH. In brief, this technique utilizes a trans-sulcal, stereotactic-guided technique in which a specially designed cannula is used to introduce a 13.5-mm-diameter tube into the epicenter of the tICH. We identified eight tICHs that were treated entirely or in part with the modified Mi SPACE technique during the time period from August 15, 2014 to December 15, 2014. This modified technique was readily deployed safely and efficaciously with significant removal of the tICH as demonstrated by postoperative CT scans. The removal of tICH using this minimally invasive technique may help with the control of ICP and cerebral edema.
Collapse
Affiliation(s)
- Jeff W Chen
- Department of Neurological Surgery, The University of California, Irvine, CA, USA.
| | - Michelle R Paff
- Department of Neurological Surgery, The University of California, Irvine, CA, USA
| | | | - Sean W Kaloostian
- Department of Neurological Surgery, The University of California, Irvine, CA, USA
| |
Collapse
|
832
|
Zinc Protoporphyrin Attenuates White Matter Injury after Intracerebral Hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:199-202. [PMID: 26463948 DOI: 10.1007/978-3-319-18497-5_35] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Intracerebral hemorrhage (ICH)-induced white matter injury has not been well studied. The objective of this study was to examine the effect of zinc protoporphyrin (ZnPP) on white matter injury induced by ICH. This study was divided into two parts. In the first part, rats received either a needle insertion (sham) or 100 μl autologous blood into the right basal ganglia. The rats were euthanized at 1, 3, 7, 14, or 28 days later for myelin basic protein (MBP) measurement. In the second part, rats had intracerebral infusion of 100 μl autologous blood, and an intraperitoneal osmotic mini-pump was implanted immediately after ICH to deliver vehicle or ZnPP (1 nmol/h), a heme oxygenase inhibitor, for up to 14 days. Rats were euthanized at day 28 for MBP staining. The number of MBP-labeled fiber bundles and their area were determined. The time-course showed that the white matter was lost in the ipsilateral basal ganglia from day 1 to day 28 after ICH. The number of MBP-labeled bundles and their area were significantly lower 2 weeks after ICH compared with sham-operated rats (p < 0.05). Systemic treatment with ZnPP attenuated the loss of MBP-labeled bundles (p < 0.01) and area (p < 0.01). In conclusion, marked white matter injury occurs after ICH. ZnPP reduces white matter injury, suggesting a role of heme degradation products in ICH-induced white matter damage.
Collapse
|
833
|
Abstract
Astrocyte endfeet envelop the cerebral capillaries that form the blood-brain barrier. Swelling of these endfeet occurs early in cerebral ischemia. It is generally hypothesized that such swelling occurs as the result of factors released from parenchymal brain cells during an ischemic stroke (e.g., K(+) and L-glutamate). In this review of mechanisms that can elicit astrocyte swelling in ischemic stroke, we hypothesize that, instead or in addition, such swelling may be a response to blood-brain barrier dysfunction. Astrocyte endfeet swelling may help form a cuff around a damaged vessel that limits the egress of plasma constituents and blood (hemorrhage) into brain.
Collapse
|
834
|
Gao F, Zheng M, Hua Y, Keep RF, Xi G. Acetazolamide Attenuates Thrombin-Induced Hydrocephalus. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:373-7. [PMID: 26463977 DOI: 10.1007/978-3-319-18497-5_64] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Our previous studies demonstrated that thrombin is an important factor in brain injury after intracerebral and intraventricular hemorrhage. This study examined the effect of acetazolamide, a carbonic anhydrase inhibitor, on thrombin-induced hydrocephalus. There were two parts in this study. First, rats had an injection of either 50 μl saline or 3 U thrombin into the right lateral ventricle. Second, rats had an injection of 3 U thrombin into the right lateral ventricle and were treated with either vehicle or acetazolamide (30 mg/kg, intraperitoneally (IP)) at 1 h after thrombin infusion. Lateral ventricle volumes were measured in magnetic resonance imaging T2 images and the brains were used for histology analysis at 24 h later. Intraventricular injection of thrombin induced significantly larger ventricle volume (27.8 ± 3.7 vs 8.5 ± 1.3 mm(3), n = 6, p < 0.01) and more ventricular wall damage (the breakdown of the ependymal layer, 20.2 ± 3.1 vs 2.4 ± 0.8 %, n = 6, p < 0.01) compared with saline injection. Acetazolamide treatment (30 mg/kg, IP) markedly attenuated thrombin-induced hydrocephalus (16.1 ± 4.2 mm(3) vs 29.5 ± 5.3 mm(3), n = 6, p < 0.01). These results suggest decreasing CSF production by acetazolamide attenuated thrombin-induced hydrocephalus in rats.
Collapse
Affiliation(s)
- Feng Gao
- Department of Neurology, The 2nd Affiliate Hospital, Zhejiang University, Zhejiang, China.,Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Mingzhe Zheng
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Hua
- Department of Neurology, The 2nd Affiliate Hospital, Zhejiang University, Zhejiang, China
| | - Richard F Keep
- Department of Neurology, The 2nd Affiliate Hospital, Zhejiang University, Zhejiang, China
| | - Guohua Xi
- Department of Neurology, The 2nd Affiliate Hospital, Zhejiang University, Zhejiang, China.
| |
Collapse
|
835
|
Src Family Kinases in Brain Edema After Acute Brain Injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:185-90. [PMID: 26463946 DOI: 10.1007/978-3-319-18497-5_33] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Brain edema, the first stage of intracranial hypertension, has been associated with poor prognosis and increased mortality after acute brain injury such as ischemic stroke, intracranial hemorrhage (ICH), and traumatic brain injury (TBI). Acute brain injury often initiates release of many molecules, including glutamate, adenosine, thrombin, oxyhemoglobin, cytokines, reactive oxygen species (ROS), damage-associated molecular pattern molecules (DAMPs), and others. Most of these molecules activate Src family kinases (SFKs), a family of proto-oncogenic non-receptor tyrosine kinases, resulting in blood-brain barrier (BBB) disruption and brain edema at the acute stage after brain injury. However, SFKs also contribute to BBB self-repair and brain edema resolution in the chronic stage that follows brain injury. In this review, we summarize possible pathways through which SFKs are implicated in both brain edema formation and its eventual resolution.
Collapse
|
836
|
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
|
837
|
Mechanisms of Cerebral Hemorrhage. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
838
|
Oxidative Stress in Intracerebral Hemorrhage: Sources, Mechanisms, and Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3215391. [PMID: 26843907 PMCID: PMC4710930 DOI: 10.1155/2016/3215391] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is associated with the highest mortality and morbidity despite only constituting approximately 10–15% of all strokes. Complex underlying mechanisms consisting of cytotoxic, excitotoxic, and inflammatory effects of intraparenchymal blood are responsible for its highly damaging effects. Oxidative stress (OS) also plays an important role in brain injury after ICH but attracts less attention than other factors. Increasing evidence has demonstrated that the metabolite axis of hemoglobin-heme-iron is the key contributor to oxidative brain damage after ICH, although other factors, such as neuroinflammation and prooxidases, are involved. This review will discuss the sources, possible molecular mechanisms, and potential therapeutic targets of OS in ICH.
Collapse
|
839
|
Xiong L, Reijmer YD, Charidimou A, Cordonnier C, Viswanathan A. Intracerebral hemorrhage and cognitive impairment. Biochim Biophys Acta Mol Basis Dis 2015; 1862:939-44. [PMID: 26692171 DOI: 10.1016/j.bbadis.2015.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/05/2015] [Accepted: 12/10/2015] [Indexed: 02/01/2023]
Abstract
Vascular cognitive impairment and vascular dementia are composed of cognitive deficits resulted from a range of vascular lesions and pathologies, including both ischemic and hemorrhagic. However the contribution of spontaneous intracerebral hemorrhage presumed due to small vessel diseases on cognitive impairment is underestimated, in contrast to the numerous studies about the role of ischemic vascular disorders on cognition. In this review we summarize recent findings from clinical studies and appropriate basic science research to better elucidate the role and possible mechanisms of intracerebral hemorrhage in cognitive impairment and dementia. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Li Xiong
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Neurology Department, Zhongnan Hospital, Wuhan University, Wuhan 430071, China.
| | - Yael D Reijmer
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Andreas Charidimou
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Charlotte Cordonnier
- Department of Neurology and Stroke Unit, Hôpital Roger Salengro, Lille University Hospital, Lille Cedex 59037, France.
| | - Anand Viswanathan
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
840
|
Klebe D, McBride D, Flores JJ, Zhang JH, Tang J. Modulating the Immune Response Towards a Neuroregenerative Peri-injury Milieu After Cerebral Hemorrhage. J Neuroimmune Pharmacol 2015; 10:576-86. [PMID: 25946986 PMCID: PMC4636976 DOI: 10.1007/s11481-015-9613-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/29/2015] [Indexed: 01/06/2023]
Abstract
Cerebral hemorrhages account for 15-20 % of stroke sub-types and have very poor prognoses. The mortality rate for cerebral hemorrhage patients is between 40 and 50 %, of which at least half of the deaths occur within the first 2 days, and 75 % of survivors are incapable of living independently after 1 year. Current emergency interventions involve lowering blood pressure and reducing intracranial pressure by controlled ventilations or, in the worst case scenarios, surgical intervention. Some hemostatic and coagulatherapeutic interventions are being investigated, although a few that were promising in experimental studies have failed in clinical trials. No significant immunomodulatory intervention, however, exists for clinical management of cerebral hemorrhage. The inflammatory response following cerebral hemorrhage is particularly harmful in the acute stage because blood-brain barrier disruption is amplified and surrounding tissue is destroyed by secreted proteases and reactive oxygen species from infiltrated leukocytes. In this review, we discuss both the destructive and regenerative roles the immune response play following cerebral hemorrhage and focus on microglia, macrophages, and T-lymphocytes as the primary agents directing the response. Microglia, macrophages, and T-lymphocytes each have sub-types that significantly influence the over-arching immune response towards either a pro-inflammatory, destructive, or an anti-inflammatory, regenerative, state. Both pre-clinical and clinical studies of cerebral hemorrhages that selectively target these immune cells are reviewed and we suggest immunomodulatory therapies that reduce inflammation, while augmenting neural repair, will improve overall cerebral hemorrhage outcomes.
Collapse
Affiliation(s)
- Damon Klebe
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Devin McBride
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jerry J Flores
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
- Departments of Anesthesiology and Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
841
|
Abstract
Intracerebral hemorrhage (ICH) is a stroke subtype with high mortality and significant disability among survivors. The management of ICH has been influenced by the results of several major trials completed in the last decade. It is now recognized that hematoma expansion is a major cause of morbidity and mortality. However, efforts to improve clinical outcome through mitigation of hematoma expansion have so far been unsuccessful. Acute blood pressure management has recently been shown to be safe in the setting of acute ICH but there was no reduction in mortality with early blood pressure (BP) lowering. Two large trials of surgical evacuation of supratentorial ICH have not shown improvement in outcome with surgery, thus minimally invasive surgical strategies are currently being studied. Lastly, a better understanding of the pathophysiology of ICH has led to the identification of several new mechanisms of injury that could be potential therapeutic targets.
Collapse
Affiliation(s)
- Venkatesh Aiyagari
- a Department of Neurological Surgery and Neurology and Neurotherapeutics , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
842
|
Yang Y, Zhang Y, Wang Z, Wang S, Gao M, Xu R, Liang C, Zhang H. Attenuation of Acute Phase Injury in Rat Intracranial Hemorrhage by Cerebrolysin that Inhibits Brain Edema and Inflammatory Response. Neurochem Res 2015; 41:748-57. [DOI: 10.1007/s11064-015-1745-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/18/2015] [Accepted: 10/22/2015] [Indexed: 11/28/2022]
|
843
|
Li D, Lei C, Zhang S, Zhang S, Liu M, Wu B. Blockade of high mobility group box-1 signaling via the receptor for advanced glycation end-products ameliorates inflammatory damage after acute intracerebral hemorrhage. Neurosci Lett 2015; 609:109-19. [PMID: 26483322 DOI: 10.1016/j.neulet.2015.10.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/15/2015] [Accepted: 10/12/2015] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating disease with no specific treatment. Increasing evidence indicates that inflammatory response plays a critical role in ICH-induced damage. High mobility group box-1 protein (HMGB1) may trigger inflammatory response via three putative receptors: receptor for advanced glycation end-products (RAGE), toll-like receptor-2 (TLR2) and toll-like receptor-4 (TLR4). Which receptor participates in HMGB1-induced inflammation during acute ICH is unknown. Using a rat model to examine the early phase of injury in collagenase-induced ICH, we found that treating animals with HMGB1 antagonist significantly reduced the expression of all three receptors. Treating animals with the HMGB1 antagonist EP or RAGE antagonist FPS-ZM1 significantly reduced inflammatory cell infiltration and expression of IL-1β, matrix metalloproteinase-9 in the perihematoma after ICH. Treatment with EP or FPS-ZM1 also led to greater neurobehavioral function and less brain edema, hemorrhage volume and brain damage after ICH. In contrast, treatment with TLR2/4 antagonists did not significantly affect these post-ICH outcomes. Our results suggest that RAGE may play a specific role in the acute phase of ICH, so targeting the HMGB1-RAGE signaling pathway may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Dan Li
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China.
| | - Chunyan Lei
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China.
| | - Shuting Zhang
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China.
| | - Shihong Zhang
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy, Ministry of Education, West China Hospital, Sichuan University, PR China.
| | - Ming Liu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy, Ministry of Education, West China Hospital, Sichuan University, PR China.
| | - Bo Wu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy, Ministry of Education, West China Hospital, Sichuan University, PR China.
| |
Collapse
|
844
|
Feng L, Chen Y, Ding R, Fu Z, Yang S, Deng X, Zeng J. P2X7R blockade prevents NLRP3 inflammasome activation and brain injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite. J Neuroinflammation 2015; 12:190. [PMID: 26475134 PMCID: PMC4609067 DOI: 10.1186/s12974-015-0409-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/05/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The NLR family, pyrin domain-containing 3 (NLRP3) inflammasome plays a key role in intracerebral hemorrhage (ICH)-induced inflammatory injury, and the purinergic 2X7 receptor (P2X7R) is upstream of NLRP3 activation. This study aimed to investigate how P2X7R functions in ICH-induced inflammatory injury and how the receptor interacts with the NLRP3 inflammasome. METHODS Rats were treated with P2X7R small interfering RNA (siRNA) 24 h before undergoing collagenase-induced ICH. A selective P2X7R inhibitor (blue brilliant G, BBG) or a peroxynitrite (ONOO(-)) decomposition catalyst (5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron(III) [FeTPPS]) was injected 30 min after ICH. Brain water content, hemorrhagic lesion volume, and neurological deficits were evaluated, and western blot, immunofluorescence, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) were carried out. RESULTS Striatal P2X7R and NLRP3 inflammasomes were activated after ICH. Gene silencing of P2X7R suppressed NLRP3 inflammasome activation and interleukin (IL)-1β/IL-18 release and significantly ameliorated brain edema and neurological deficits. Additionally, enhanced NADPH oxidase 2 (NOX2, gp91(phox)) and inducible nitric oxide synthase (iNOS), as well as their cytotoxic product (ONOO(-)) were markedly attenuated by BBG treatment following ICH. This was accompanied by downregulations of the inflammasome components, IL-1β/IL-18 and myeloperoxidase (MPO, a neutrophil marker). Most importantly, inflammasome activation and IL-1β/IL-18 release were significantly inhibited by ONOO(-) decomposition with FeTPPS. CONCLUSIONS Our findings implicate that P2X7R exacerbated inflammatory progression and brain damage in ICH rats possibly via NLRP3 inflammasome-dependent IL-1β/IL-18 release and neutrophil infiltration. ONOO(-), a potential downstream signaling molecule of P2X7R, may play a critical role in triggering NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Liang Feng
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Yizhao Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Rui Ding
- Department of Neurosurgery, Jingmen No. 1 People's Hospital, Jingmen, 448000, Hubei, China.
| | - Zhenghao Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, China.
| | - Shuo Yang
- Department of Neurosurgery, Gaoqing Campus of Central Hospital of Zibo, Gaoqing People's Hospital, Gaoqing, Zibo, 256300, Shandong, China.
| | - Xinqing Deng
- Department of Neurosurgery, 999 Brain Hospital, Jinan University, Guangzhou, 510510, Guangdong, China.
| | - Jun Zeng
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
845
|
Sheth KN, Martini SR, Moomaw CJ, Koch S, Elkind MSV, Sung G, Kittner SJ, Frankel M, Rosand J, Langefeld CD, Comeau ME, Waddy SP, Osborne J, Woo D. Prophylactic Antiepileptic Drug Use and Outcome in the Ethnic/Racial Variations of Intracerebral Hemorrhage Study. Stroke 2015; 46:3532-5. [PMID: 26470777 DOI: 10.1161/strokeaha.115.010875] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/16/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE The role of antiepileptic drug (AED) prophylaxis after intracerebral hemorrhage (ICH) remains unclear. This analysis describes prevalence of prophylactic AED use, as directed by treating clinicians, in a prospective ICH cohort and tests the hypothesis that it is associated with poor outcome. METHODS Analysis included 744 patients with ICH enrolled in the Ethnic/Racial Variations of Intracerebral Hemorrhage (ERICH) study before November 2012. Baseline clinical characteristics and AED use were recorded in standardized fashion. ICH location and volume were recorded from baseline neuroimaging. We analyzed differences in patient characteristics by AED prophylaxis, and we used logistic regression to test whether AED prophylaxis was associated with poor outcome. The primary outcome was 3-month modified Rankin Scale score, with 4 to 6 considered poor outcome. RESULTS AEDs were used for prophylaxis in 289 (39%) of the 744 subjects; of these, levetiracetam was used in 89%. Patients with lobar ICH, craniotomy, or larger hematomas were more likely to receive prophlyaxis. Although prophylactic AED use was associated with poor outcome in an unadjusted model (odds ratio, 1.40; 95% confidence interval, 1.04-1.88; P=0.03), this association was no longer significant after adjusting for clinical and demographic characteristics (odds ratio, 1.11; 95% confidence interval, 0.74-1.65; P=0.62). CONCLUSIONS We found no evidence that AED use (predominantly levetiracetam) is independently associated with poor outcome. A prospective study is required to assess for a more modest effect of AED use on outcome after ICH.
Collapse
Affiliation(s)
- Kevin N Sheth
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.).
| | - Sharyl R Martini
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Charles J Moomaw
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Sebastian Koch
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Mitchell S V Elkind
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Gene Sung
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Steven J Kittner
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Michael Frankel
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Jonathan Rosand
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Carl D Langefeld
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Mary E Comeau
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Salina P Waddy
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Jennifer Osborne
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | - Daniel Woo
- From the Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); Department of Neurology, Baylor College of Medicine, Houston, TX (S.R.M.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., J.O., D.W.); Department of Neurology, University of Miami School of Medicine, Miami, FL (S.K.); Department of Neurology, College of Physicians and Surgeons, Columbia University, New York (M.S.V.E.); Department of Neurology, University of Southern California, Los Angeles (G.S.); Department of Neurology, University of Maryland School of Medicine, Baltimore, MD (S.J.K.); Department of Neurology, Emory University School of Medicine, Atlanta, GA (M.F.); Department of Neurology, Massachusetts General Hospital, Boston, MA (J.R.); Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC (C.D.L., M.E.C.); and Office of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD (S.P.W.)
| | | |
Collapse
|
846
|
Majidi S, Rahim B, Gilani SI, Gilani WI, Adil MM, Qureshi AI. CT Evolution of Hematoma and Surrounding Hypodensity in a Cadaveric Model of Intracerebral Hemorrhage. J Neuroimaging 2015; 26:346-50. [DOI: 10.1111/jon.12306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/03/2015] [Accepted: 08/31/2015] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shahram Majidi
- Zeenat Qureshi Stroke Institute; St Cloud; MN
- Department of Neurology; George Washington University; Washington D.C
| | - Basit Rahim
- Zeenat Qureshi Stroke Institute; St Cloud; MN
| | | | | | | | | |
Collapse
|
847
|
Zhao H, Garton T, Keep RF, Hua Y, Xi G. Microglia/Macrophage Polarization After Experimental Intracerebral Hemorrhage. Transl Stroke Res 2015; 6:407-9. [PMID: 26446073 DOI: 10.1007/s12975-015-0428-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/27/2015] [Indexed: 12/01/2022]
Affiliation(s)
- Hao Zhao
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Thomas Garton
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
848
|
Murthy SB, Moradiya Y, Dawson J, Lees KR, Hanley DF, Ziai WC. Perihematomal Edema and Functional Outcomes in Intracerebral Hemorrhage: Influence of Hematoma Volume and Location. Stroke 2015; 46:3088-92. [PMID: 26396030 DOI: 10.1161/strokeaha.115.010054] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/26/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE Perihematomal edema (PHE) is associated with poor outcomes after intracerebral hemorrhage (ICH). PHE evolves in the early period after ICH, providing a therapeutic target and window for intervention. We studied the effect of PHE volume expansion in the first 72 hours (iPHE) and its relationship with functional outcomes. METHODS We used data contained in the Virtual International Stroke Trials Archive. We included patients who presented within 6 hours of symptom onset, had baseline clinical, radiological, and laboratory data, and further computed tomographic scan data at 72 hours and 90-day functional outcomes. We calculated iPHE and used logistic regression analysis to assess relationships with outcome. We adjusted for confounding variables and the primary outcome measure poor day-90 outcome (defined as modified Rankin Scale score of ≥3. We performed subgroup analyses by location and by volume of ICH. RESULTS We included 596 patients with ICH. Median baseline hematoma volume was 15.0 mL (IQR, 7.9-29.2) and median baseline PHE volume was 8.7 mL (IQR, 4.5-15.5). Hematoma expansion occurred in 122 (34.9%) patients. Median iPHE was 14.7 mL (IQR, 6.6-30.3). The odds of a poor outcome were greater with increasing iPHE (OR, 1.78; CI, 1.12-2.64 per mL increase). Subgroup analyses showed that iPHE was only related to poor functional outcomes in basal ganglia and small (<30 mL) ICH. CONCLUSIONS Absolute increase in PHE during 72 hours was associated with worse functional outcomes after ICH, particularly with basal ganglia ICH and hematomas <30 mL.
Collapse
Affiliation(s)
- Santosh B Murthy
- From the Division of Neurosciences Critical Care, Department of Neurology (S.B.M., Y.M., W.C.Z.) and Division of Brain Injury Outcomes (D.F.H.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Cerebrovascular Medicine, University of Glasgow, United Kingdom (J.D., K.R.L.).
| | - Yogesh Moradiya
- From the Division of Neurosciences Critical Care, Department of Neurology (S.B.M., Y.M., W.C.Z.) and Division of Brain Injury Outcomes (D.F.H.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Cerebrovascular Medicine, University of Glasgow, United Kingdom (J.D., K.R.L.)
| | - Jesse Dawson
- From the Division of Neurosciences Critical Care, Department of Neurology (S.B.M., Y.M., W.C.Z.) and Division of Brain Injury Outcomes (D.F.H.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Cerebrovascular Medicine, University of Glasgow, United Kingdom (J.D., K.R.L.)
| | - Kennedy R Lees
- From the Division of Neurosciences Critical Care, Department of Neurology (S.B.M., Y.M., W.C.Z.) and Division of Brain Injury Outcomes (D.F.H.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Cerebrovascular Medicine, University of Glasgow, United Kingdom (J.D., K.R.L.)
| | - Daniel F Hanley
- From the Division of Neurosciences Critical Care, Department of Neurology (S.B.M., Y.M., W.C.Z.) and Division of Brain Injury Outcomes (D.F.H.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Cerebrovascular Medicine, University of Glasgow, United Kingdom (J.D., K.R.L.)
| | - Wendy C Ziai
- From the Division of Neurosciences Critical Care, Department of Neurology (S.B.M., Y.M., W.C.Z.) and Division of Brain Injury Outcomes (D.F.H.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Cerebrovascular Medicine, University of Glasgow, United Kingdom (J.D., K.R.L.)
| | | |
Collapse
|
849
|
Impact of Perihemorrhagic Edema on Short-Term Outcome After Intracerebral Hemorrhage. Neurocrit Care 2015; 24:404-12. [DOI: 10.1007/s12028-015-0185-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
850
|
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
|