851
|
Sukumari-Ramesh S, Alleyne CH, Dhandapani KM. The Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid (SAHA) Confers Acute Neuroprotection After Intracerebral Hemorrhage in Mice. Transl Stroke Res 2015; 7:141-8. [PMID: 26338677 DOI: 10.1007/s12975-015-0421-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 01/19/2023]
Abstract
Spontaneous intracerebral hemorrhage (ICH) is a stroke subtype with no effective treatment. Though ICH is known to induce severe neurological damage, the molecular mechanisms of neurological injury after ICH remain largely unclear. Given the emerging role of epigenetic mechanisms in neurodegeneration, the present study evaluated whether suberoylanilide hydroxamic acid (SAHA: vorinostat), a clinically well-tolerated pan-histone deacetylase inhibitor (HDACi), would attenuate neurological injury and improve functional outcomes in a preclinical model of ICH. Mice were administered with SAHA or vehicle after an induction of ICH and acute neuronal death, glial activation, and neurological outcomes were assessed. SAHA-treated mice exhibited less neurodegeneration with concomitant improvement in neurological outcomes than vehicle-treated mice. Furthermore, SAHA downregulated glial activation and the expression of heme oxygenase-1, a stress-inducible enzyme that plays critical roles in neurological damage after ICH. Altogether, the data strongly suggest the role of epigenetic mechanisms in inducing neurological injury after ICH and raise the possible clinical utility of SAHA for therapeutic intervention after ICH.
Collapse
Affiliation(s)
- Sangeetha Sukumari-Ramesh
- Department of Neurosurgery, Georgia Regents University, 1120 15th Street CA1010, Augusta, GA, 30912, USA.
| | - Cargill H Alleyne
- Department of Neurosurgery, Georgia Regents University, 1120 15th Street CA1010, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Georgia Regents University, 1120 15th Street CA1010, Augusta, GA, 30912, USA
| |
Collapse
|
852
|
Xu Z, Li X, Chen J, Zhao J, Wang J, Ji Y, Shen Y, Han L, Shi J, Zhang D. USP11, Deubiquitinating Enzyme, Associated with Neuronal Apoptosis Following Intracerebral Hemorrhage. J Mol Neurosci 2015; 58:16-27. [PMID: 26334325 DOI: 10.1007/s12031-015-0644-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 08/18/2015] [Indexed: 11/29/2022]
Abstract
Protein ubiquitination is a dynamic two-way process that can be reversed or regulated by deubiquitinating enzymes (DUB). USP11, located on the X chromosome, 6 is a member of USP subclass of the DUB family. Here, we demonstrate that USP11 may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). From the results of Western blot, immunohistochemistry, and immunofluorescence, we obtained a significant up-regulation of USP11 in neurons adjacent to the hematoma following ICH. Increasing USP11 level was found to be accompanied by the up-regulation of active caspase-3, Fas receptor (Fas), Fas ligand (FasL), and active caspase-8. Besides, USP11 co-localized well with active caspase-3 in neurons, indicating its potential role in neuronal apoptosis. What is more, knocking down USP11 by RNA-interference in PC12 cells reduced active caspase-3 expression. Thus, USP11 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 Province, China
| | - Xiaohong Li
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jianping Chen
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jianmei Zhao
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jun Wang
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, 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 Province, China
| | - Yifen Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Lijian Han
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Jiansheng Shi
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, 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 Province, China.
| |
Collapse
|
853
|
Role of lipocalin-2 in brain injury after intracerebral hemorrhage. J Cereb Blood Flow Metab 2015; 35:1454-61. [PMID: 25853903 PMCID: PMC4640334 DOI: 10.1038/jcbfm.2015.52] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/24/2015] [Accepted: 03/02/2015] [Indexed: 01/08/2023]
Abstract
Lipocalin-2 (LCN2) is a siderophore-binding protein involved in cellular iron transport and neuroinflammation. Both iron and inflammation are involved in brain injury after intracerebral hemorrhage (ICH) and this study examined the role of LCN2 in such injury. Male adult C57BL/6 wild-type (WT) or LCN2-deficient (LCN2(-/-)) mice had an intracerebral injection of autologous blood or FeCl2. Control animals had a sham operation or saline injection. T2-weighted magnetic resonance imaging and behavioral tests were performed at days 1, 3, 7, 14, and 28 after injection. In WT mice, brain LCN2 levels were increased in the ipsilateral basal ganglia after ICH or iron injection. Lipocalin-2-positive cells were astrocytes, microglia, neurons, and endothelial cells. Intracerebral hemorrhage resulted in a significant increase in ferritin expression in the ipsilateral basal ganglia. Compared with WT mice, ICH caused less ferritin upregulation, microglia activation, brain swelling, brain atrophy, and neurologic deficits in LCN2(-/-) mice (P<0.05). The size of the lesion induced by FeCl2 injection as well as the degree of brain swelling and blood-brain barrier disruption were also less in LCN2(-/-) mice (P<0.05). These results suggest a role of LCN2 in enhancing brain injury and iron toxicity after ICH.
Collapse
|
854
|
Abstract
PURPOSE OF REVIEW Intracerebral haemorrhage is a devastating cerebrovascular disease with no established treatment. Its course is often complicated by secondary haematoma expansion and perihemorrhagic oedema. Decompressive hemicraniectomy is effective in the treatment of space-occupying hemispheric ischaemic stroke. The purpose of this review is to assess the role of decompressive hemicraniectomy in intracerebral haemorrhage. RECENT FINDINGS After few small previous studies had suggested advantages by the combination of decompressive hemicraniectomy with haematoma removal, decompression on its own has been investigated within the last 5 years. Two case series and one case-control study in altogether 40 patients with severe spontaneous intracerebral haemorrhage have shown mortality rates ranging from 13 to 25% and favourable outcome from 40 to 65%. SUMMARY Decompressive hemicraniectomy appears to be a feasible and relatively well tolerated individual treatment option for selected patients with spontaneous intracerebral haemorrhage. Data are insufficient to judge potential benefits in outcome. A randomized trial is justified and mandatory.
Collapse
|
855
|
Park HK, Chon J, Park HJ, Chung JH, Baik HH. Association between two promoter polymorphisms (rs1893219 and rs1893220) of MC2R gene and intracerebral hemorrhage in Korean population. Neurosci Lett 2015; 602:1-5. [PMID: 26115626 DOI: 10.1016/j.neulet.2015.06.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/01/2015] [Accepted: 06/16/2015] [Indexed: 11/26/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis has an important role in the pathogenesis of stroke. We investigated whether single nucleotide polymorphisms (SNPs) of melanocortin 2 receptor (MC2R), also known as adrenocorticotropic hormone (ACTH) receptor, were associated with the development of intracerebral hemorrhage (ICH) in Korean population. Two promoter SNPs [rs1893219 (-853A/G) and rs1893220 (-759G/T)] were genotyped in 145 ICH patients and 331 control subjects using direct sequencing. Multiple logistic regression models were used to determine odds ratios, 95% confidence intervals, and p-values. Two SNPs were associated with the development of ICH (rs1893219, p=0.003 in log-additive model, p=0.023 in dominant model, p=0.002 in recessive model; rs1893220, p=0.005 in log-additive model, p=0.021 in dominant model, p=0.003 in recessive model). The frequencies of the G allele of rs1893219 and the T allele of rs1893220 were decreased in ICH group compared to control group (p=0.003 and p=0.004, respectively). The frequencies of the AG and GT haplotypes comprised of rs1893219 and rs1893220 were also significantly different between the ICH and control groups (p=0.0026 and p=0.0034, respectively). These data suggest that the MC2R gene may contribute to the development of ICH.
Collapse
Affiliation(s)
- Hyun-Kyung Park
- Department of Emergency Medicine, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinmann Chon
- Department of Physical Medicine and Rehabilitation, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hae Jeong Park
- Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Joo-Ho Chung
- Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Hyung Hwan Baik
- Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
856
|
Chen Q, Tang J, Tan L, Guo J, Tao Y, Li L, Chen Y, Liu X, Zhang JH, Chen Z, Feng H. Intracerebral Hematoma Contributes to Hydrocephalus After Intraventricular Hemorrhage via Aggravating Iron Accumulation. Stroke 2015; 46:2902-8. [PMID: 26265129 DOI: 10.1161/strokeaha.115.009713] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/14/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE The intraventricular hemorrhage (IVH) secondary to intracerebral hemorrhage (ICH) was reported to be relevant to a higher incidence of hydrocephalus, which would result in poorer outcomes for patients with ICH. However, the mechanisms responsible for this relationship remain poorly characterized. Thus, this study was designed to further explore the development and progression of hydrocephalus after secondary IVH. METHODS Autologous blood injection model was induced to mimic ICH with ventricular extension (ICH/IVH) or primary IVH in Sprague-Dawley rats. Magnetic resonance imaging, Morris water maze, brain water content, Evans blue extravasation, immunohistochemistry staining, Western blot, iron determination, and electron microscopy were used in these rats. Then, deferoxamine treatment was used to clarify the involvement of iron in the development of hydrocephalus. RESULTS Despite the injection of equivalent blood volumes, ICH/IVH resulted in more significant ventricular dilation, ependymal cilia damage, and iron overload, as well as more severe early brain injury and neurological deficits compared with IVH alone. Systemic deferoxamine treatment more effectively reduced ventricular enlargement in ICH/IVH compared with primary IVH. CONCLUSIONS Our results show that ICH/IVH caused more significant chronic hydrocephalus and iron accumulation than primary IVH alone. Intracerebral hematoma plays a vital role in persistent iron overload and aggravated hydrocephalus after ICH/IVH.
Collapse
Affiliation(s)
- Qianwei Chen
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Jun Tang
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Liang Tan
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Jing Guo
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Yihao Tao
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Lin Li
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Yujie Chen
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Xin Liu
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - John H Zhang
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.)
| | - Zhi Chen
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.).
| | - Hua Feng
- From the Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China (Q.C., J.T., L.T., J.G., Y.T., L.L., Y.C., X.L., Z.C., H.F.); and Department of Anesthesia, Neurosurgery and Physiology, Loma Linda University, CA (J.H.Z.).
| |
Collapse
|
857
|
Mu HM, Wang LY. Effect of therapeutic ultrasound on brain angiogenesis following intracerebral hemorrhage in rats. Microvasc Res 2015; 102:11-8. [PMID: 26265191 DOI: 10.1016/j.mvr.2015.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/04/2015] [Accepted: 08/04/2015] [Indexed: 11/29/2022]
Abstract
Intracerebral hemorrhage (ICH) can produce severe neurological deficits in stroke survivors. However, few effective approaches are available to improve the recovery from ICH. Given that therapeutic ultrasound exposure can enhance on angiogenesis in peripheral tissues, the present study was designed to examine the effects of therapeutic ultrasound exposure on the brain angiogenesis following ICH. To this end, we applied once daily therapeutic ultrasound treatment to rats for 7 consecutive days after intracranial infusion of vehicle (Sham control) or collagenase (ICH). Repeated exposure to the low intensity of therapeutic ultrasound decreased behavioral scores in ICH rats, but not in sham control rats. Such an effect was correlated with an increased number of vessel-like structures and microvessels and PCNA positive cells in vWF-positive blood vessels in perihematomal brain tissues at post-ICH day 7. Furthermore, immunohistochemistry and western blotting results showed that ICH trigged the expression of extracellular matrix (ECM)-related molecules, including collagen Is, III, and IV, as well as integrins αvβ3 and α5β1, and exposure to therapeutic ultrasound increased the expression of these molecules. Therefore, our results indicated that repeated exposure to a low intensity of therapeutic ultrasound can increase the expression of collagen and integrins of ECM-related molecules, promote the formation of a large number of vessel-like structure and capillaries around the hematoma, and accelerate the recovery of neurological function impaired by ICH.
Collapse
Affiliation(s)
- Hong-Mei Mu
- Department of Ultrasonography, Cangzhou Central Hospital, Cangzhou 061000, Hebei, China
| | - Li-Yong Wang
- Department of Neurology, Cangzhou People's Hospital, Cangzhou 061000, Hebei, China.
| |
Collapse
|
858
|
Zhang Y, Chen Y, Wu J, Manaenko A, Yang P, Tang J, Fu W, Zhang JH. Activation of Dopamine D2 Receptor Suppresses Neuroinflammation Through αB-Crystalline by Inhibition of NF-κB Nuclear Translocation in Experimental ICH Mice Model. Stroke 2015; 46:2637-46. [PMID: 26251254 DOI: 10.1161/strokeaha.115.009792] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/06/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Inflammatory injury plays a critical role in intracerebral hemorrhage (ICH)-induced secondary brain injury. Recently, dopamine D2 receptor (DRD2) is identified as an important component controlling innate immunity and inflammatory response in central nervous system, and αB-crystallin (CRYAB) is a potent negative regulator on inflammatory pathways. Here, we sought to investigate the role of DRD2 on neuroinflammation after experimental ICH and the potential mechanism mediated by CRYAB. METHODS Two hundred and twenty-four (224) male CD-1 mice were subjected to intrastriatal infusion of bacterial collagenase or autologous blood. Two DRD2 agonists quinpirole and ropinirole were administrated by daily intraperitoneal injection starting at 1 hour after ICH. DRD2 and CRYAB in vivo knockdown was performed 48 hours before ICH insult. Behavioral deficits and brain water content, Western blots, immunofluorescence staining, coimmunoprecipitation (Co-IP) assay, and proteome cytokine array were evaluated. RESULTS Endogenous DRD2 and CRYAB expressions were increased after ICH. DRD2 knockdown aggravated the neurobehavioral deficits and the pronounced cytokine expressions. DRD2 activation by quinpirole and ropinirole ameliorated neurological outcome, brain edema, interleukin-1β, and monocyte chemoattractant protein-1 expression, as well as microglia/macrophages activation, in the perihematomal region. These effects were abolished by pretreatment with CRYAB siRNAs. Quinpirole enhanced cytoplasmic binding activity between CRYAB and NF-κB and decreased nuclear NF-κB expression. Similar therapeutic benefits were observed using autologous blood injection model and intranasal delivery of quinpirole. CONCLUSIONS DRD2 may have anti-inflammatory effects after ICH. DRD2 agonists inhibited neuroinflammation and attenuated brain injury after ICH, which is probably mediated by CRYAB and enhanced cytoplasmic binding activity with NF-κB.
Collapse
Affiliation(s)
- Yang Zhang
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.)
| | - Yujie Chen
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.)
| | - Jiang Wu
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.)
| | - Anatol Manaenko
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.)
| | - Peng Yang
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.)
| | - Jiping Tang
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.)
| | - Weiling Fu
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.).
| | - John H Zhang
- From the Department of Laboratory Medicine (Y.Z., W.F.) and Department of Neurosurgery (Y.C.), Southwest Hospital, Third Military Medical University, Chongqing, China; Department of Physiology and Pharmacology, Loma Linda University, CA (Y.Z., Y.C., J.W., A.M., P.Y., J.T., J.H.Z.); and Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China (J.W.).
| |
Collapse
|
859
|
Iniaghe LO, Krafft PR, Klebe DW, Omogbai EKI, Zhang JH, Tang J. Dimethyl fumarate confers neuroprotection by casein kinase 2 phosphorylation of Nrf2 in murine intracerebral hemorrhage. Neurobiol Dis 2015; 82:349-358. [PMID: 26176793 DOI: 10.1016/j.nbd.2015.07.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/29/2015] [Accepted: 07/04/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Edema formation, inflammation and increased blood-brain barrier permeability contribute to poor outcomes after intracerebral hemorrhage (ICH). This study examined the therapeutic effect of dimethyl fumarate (DMF), a fumaric acid ester that activates nuclear factor erythroid-2 related factor 2 (Nrf2) and Nrf2 heterodimerization effector protein musculo-aponeurotic fibrosarcoma-G (MAFG) in a murine ICH model. METHODS Male CD-1 mice (n=176) were subjected to intrastriatal infusion of bacterial collagenase (n=126), autologous blood (n=18) or sham surgery (n=32). Four (4) animals not subjected to ICH (naive) were also included in the study. After ICH, animals either received vehicle, dimethyl fumarate (10 mg or 100 mg/kg) or casein kinase 2 inhibitor (E)-3-(2,3,4,5-tetrabromophenyl)acrylic acid (TBCA). Thirty-two mice also received scrambled siRNA or MAFG siRNA 24h before ICH. Brain water content and neurological function were evaluated. RESULTS Dimethyl fumarate reduced Evans blue dye extravasation, decreased brain water content, and improved neurological deficits at 24 and 72 h after ICH. Casein kinase 2 inhibitor TBCA and MAFG siRNA prevented the effect of dimethyl fumarate on brain edema and neurological function. After ICH, ICAM-1 levels increased and casein kinase 2 levels decreased. Dimethyl fumarate reduced ICAM-1 but enhanced casein kinase 2 levels. Again, casein kinase 2 inhibitor TBCA and MAFG siRNA abolished the effect of dimethyl fumarate on ICAM-1 and casein kinase 2. Dimethyl fumarate preserved pNrf2 and MAFG expression in the nuclear lysate after ICH and the effect of dimethyl fumarate was abolished by casein kinase 2 inhibitor TBCA and MAFG siRNA. Dimethyl fumarate reduced microglia activation in peri-hematoma areas after ICH. The protective effect of dimethyl fumarate on brain edema and neurological function was also observed in a blood injection mouse model. CONCLUSION Dimethyl fumarate ameliorated inflammation, reduced blood-brain barrier permeability, and improved neurological outcomes by casein kinase 2 and Nrf2 signaling pathways after experimental ICH in mice.
Collapse
Affiliation(s)
- Loretta O Iniaghe
- Department of Physiology and Pharmacology, Loma Linda University, USA; Department of Pharmacology and Toxicology, University of Benin, Nigeria
| | - Paul R Krafft
- Department of Physiology and Pharmacology, Loma Linda University, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| | - Damon W Klebe
- Department of Physiology and Pharmacology, Loma Linda University, USA
| | - Eric K I Omogbai
- Department of Pharmacology and Toxicology, University of Benin, Nigeria
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, USA; Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, USA.
| |
Collapse
|
860
|
Male-Specific Alleviation of Iron-Induced Striatal Injury by Inhibition of Autophagy. PLoS One 2015; 10:e0131224. [PMID: 26147959 PMCID: PMC4492841 DOI: 10.1371/journal.pone.0131224] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/30/2015] [Indexed: 02/03/2023] Open
Abstract
Men exhibit a worse survival rate than premenopausal women after intracerebral hemorrhage (ICH), however, no sex-specific management has been concerned. In a rat model involving infusion of ferrous citrate (FC) that simulates iron accumulation after hemorrhage, a higher degree of autophagy associated with higher injury severity was observed in striatum of males than in females. Since the imbalance between the levels of autophagy and energy demand may lead to cell death, we proposed that FC-induced autophagy is detrimental in a male specific manner and autophagy modulation affects injury severity in a sex-dependent manner. Rapamycin, an autophagy inducer, and conditional knockout gene of autophagy-related protein 7 (Atg7) in dopamine receptor D2 (DRD2) neurons were used to test our hypothesis using a mouse model with striatal FC infusion. The result showed that the levels of autophagic cell death and injury severity were higher in male than in female mice. Pre-treatment of FC-infused females with rapamycin increased the FC-induced behavioral deficit and DRD2 neuron death. However, DRD2 neuron-specific knockout of Atg7 decreased FC-induced injury severity and the number of TUNEL(+) DRD2 neurons in males. These results suggest that autophagy in FC-infusion males is overactive with maladaptive consequences and inhibition of autophagy decreases the severity of FC-induced striatal injury in males. These findings present prospects for male-specific therapeutic strategy that targets autophagy in patients suffering from iron overload.
Collapse
|
861
|
Li A, Li L, Sun X, Ni Y, Chen X, Guo A, Chen X. Increased Expression of Mitochondrial Inner-Membrane Protein Mpv17 After Intracerebral Hemorrhage in Adult Rats. Neurochem Res 2015; 40:1620-30. [PMID: 26123482 DOI: 10.1007/s11064-015-1644-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/15/2015] [Accepted: 06/17/2015] [Indexed: 01/18/2023]
Abstract
The Mpv17 gene encodes a mitochondrial inner-membrane protein that has been implicated in several cell activities. Almost all studies have previously indicated that loss of function or gene-inactivated in Mpv17 can induce the development of disease. Here, we explored the roles of Mpv17 protein in the pathophysiology of intracerebral hemorrhage (ICH). An ICH rat model was established and assessed by behavioral tests. Using western blot and immunohistochemistry, significant up-regulation of Mpv17 was found in neurons in brain areas surrounding the hematoma following ICH. The increase of Mpv17 expression was found to be accompanied by the enhanced expression of p53, Bax, cytochrome c (Cyt c) and active caspase-3, and decreased expression of Bcl-2 in the pathological process of rat ICH. Furthermore, immunofluorescent staining revealed that Mpv17 co-localized with p53, Bax and active caspase-3 in neurons, suggesting its biological function in the process of neuronal apoptosis. Our in vitro study, using Mpv17 RNA interference in primary cortical neurons, indicated that Mpv17 might exert its anti-apoptotic function in neuronal apoptosis. Thus, Mpv17 may play a role in protecting the brain from secondary damage following ICH.
Collapse
Affiliation(s)
- Aihong Li
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | | | | | | | | | | | | |
Collapse
|
862
|
Qin J, Ma X, Qi H, Song B, Wang Y, Wen X, Wang QM, Sun S, Li Y, Zhang R, Liu X, Hou H, Gong G, Xu Y. Transplantation of Induced Pluripotent Stem Cells Alleviates Cerebral Inflammation and Neural Damage in Hemorrhagic Stroke. PLoS One 2015; 10:e0129881. [PMID: 26086994 PMCID: PMC4472717 DOI: 10.1371/journal.pone.0129881] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 05/15/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Little is known about the effects of induced pluripotent stem cell (iPSC) treatment on acute cerebral inflammation and injuries after intracerebral hemorrhage (ICH), though they have shown promising therapeutic potentials in ischemic stoke. METHODS An ICH model was established by stereotactic injection of collagenase VII into the left striatum of male Sprague-Dawley (SD) rats. Six hours later, ICH rats were randomly divided into two groups and received intracerebrally 10 μl of PBS with or without 1 × 10(6) of iPSCs. Subsequently, neural function of all ICH rats was assessed at days 1, 3, 7, 14, 28 and 42 after ICH. Inflammatory cells, cytokines and neural apoptosis in the rats' perihematomal regions, and brain water content were determined on day 2 or 3 post ICH. iPSC differentiation was determined on day 28 post ICH. Nissl(+) cells and glial fibrillary acidic protein (GFAP)(+) cells in the perihematoma and the survival rates of rats in two groups were determined on post-ICH day 42. RESULTS Compared with control animals, iPSCs treatment not only improved neurological function and survival rate, but also resulted in fewer intracephalic infiltrations of neutrophils and microglia, along with decreased interleukin (IL)-1β, IL-6 and tumour necrosis factor-alpha (TNF-α), and increased IL-10 in the perihematomal tissues of ICH rats. Furthermore, brain oedema formation, apoptosis, injured neurons and glial scar formation were decreased in iPSCs-transplanted rats. CONCLUSIONS Our findings indicate that iPSCs transplantation attenuate cerebral inflammatory reactions and neural injuries after ICH, and suggests that multiple mechanisms including inflammation modulation, neuroprotection and functional recovery might be involved simultaneously in the therapeutic benefit of iPSC treatment against hemorrhagic stroke.
Collapse
Affiliation(s)
- Jie Qin
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Xun Ma
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Haiyun Qi
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Bo Song
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Yanlin Wang
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA 23284, United States of America
| | - Qing Mei Wang
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Shilei Sun
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Yusheng Li
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Rui Zhang
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Xinjing Liu
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Haiman Hou
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Guangming Gong
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
- * E-mail: (YX); (GG)
| | - Yuming Xu
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
- * E-mail: (YX); (GG)
| |
Collapse
|
863
|
Kim K, Park HW, Moon HE, Kim JW, Bae S, Chang JW, Oh W, Yang YS, Paek SH. The Effect of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells in a Collagenase-Induced Intracerebral Hemorrhage Rat Model. Exp Neurobiol 2015; 24:146-55. [PMID: 26113794 PMCID: PMC4479811 DOI: 10.5607/en.2015.24.2.146] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/04/2015] [Accepted: 05/08/2015] [Indexed: 01/01/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the devastating types of stroke. Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have potential benefits in recovery from brain damage following ICH. This study aimed to identify the beneficial effects of hUCB-MSCs and investigate whether they have anti-inflammatory effects on the ICH brain via neurotrophic factors or cytokines. hUCB-MSCs were transplanted into a collagenase-induced ICH rat model. At 2, 9, 16, and 30 days after ICH, rotarod and limb placement tests were performed to measure behavioral outcomes. ICH rats were sacrificed to evaluate the volume of lesion using H&E staining. Immunostaining was performed to investigate neurogenesis, angiogenesis, and anti-apoptosis at 4 weeks after transplantation. Inflammatory factors (TNF-α, COX-2, microglia, and neutrophils) were analyzed by immunofluorescence staining, RT-PCR, and Western blot at 3 days after transplantation. hUCB-MSCs were associated with neurological benefits and reduction in lesion volume. The hUCB-MSCs-treated group tended to reveal high levels of neurogenesis, angiogenesis, and anti-apoptosis (significant for angiogenesis). The expression levels of inflammatory factors tended to be reduced in the hUCB-MSCs-treated group compared with the controls. Our study suggests that hUCB-MSCs may improve neurological outcomes and modulate inflammation-associated immune cells and cytokines in ICH-induced inflammatory responses.
Collapse
Affiliation(s)
- Kwanwoo Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hyo-Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Seongtae Bae
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Jong Wook Chang
- Stem Cells & Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul 137-874, Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul 137-874, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| |
Collapse
|
864
|
The Pathophysiology of Intracerebral Hemorrhage Formation and Expansion. Transl Stroke Res 2015; 6:257-63. [DOI: 10.1007/s12975-015-0410-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
|
865
|
Parry-Jones AR, Wang X, Sato S, Mould WA, Vail A, Anderson CS, Hanley DF. Edema Extension Distance. Stroke 2015; 46:e137-40. [DOI: 10.1161/strokeaha.115.008818] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Adrian R. Parry-Jones
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| | - Xia Wang
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| | - Shoichiro Sato
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| | - W. Andrew Mould
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| | - Andy Vail
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| | - Craig S. Anderson
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| | - Daniel F. Hanley
- From the Centre for Vascular and Stroke Research, Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust (A.R.P.-J., A.V.) and Centre for Biostatistics (A.V.), University of Manchester, Manchester, United Kingdom; Greater Manchester Comprehensive Stroke Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (A.R.P.-J.); Neurological and Mental Health Division, The George Institute for Global Health, University of Sydney and Royal Prince Alfred Hospital, Sydney,
| |
Collapse
|
866
|
Xiong XY, Chen J, Zhu WY, Zhao T, Zhong Q, Zhou K, Meng ZY, Wang YC, Wang PF, Fang H, Yang QW. Serum hepcidin concentrations correlate with serum iron level and outcome in patients with intracerebral hemorrhage. Neurol Sci 2015; 36:1843-9. [PMID: 26024728 DOI: 10.1007/s10072-015-2266-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/22/2015] [Indexed: 12/20/2022]
Abstract
Iron plays a detrimental role in the intracerebral hemorrhage (ICH)-induced brain damage, while hepcidin is the most important iron-regulated hormone. Here, we investigate the association between serum hepcidin and serum iron, outcome in patients with ICH. Serum samples of 81 cases with ICH were obtained on consecutive days to detect the levels of hepcidin, iron, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). The National Institutes of Health Stroke Scale score (NIHSS) was measured at admission and on days 7 and 30, and the modified Rankin Scale (mRS) score was evaluated at 3 months after ICH. Additionally, the correlations of serum hepcidin with serum iron and the mRS score were analyzed by a generalized linear model. Higher serum hepcidin levels were detected in patients with poor outcomes (P < 0.001), and the mRS score increased by a mean of 1.135 points (95% CI 1.021-1.247, P < 0.001) for every serum hepcidin quartile after adjusting for other prognostic variables. Pearson correlation analysis showed that serum hepcidin was negatively correlated with serum iron (r = -0.5301, P < 0.001), and a significantly lower concentration of serum iron was found in patients with poor outcomes (P = 0.007). Additionally, serum hepcidin was independently correlated with mRS scores of ICH patients (OR 1.115, 95% CI 0.995-1.249, P = 0.021). Our results suggest that serum hepcidin is closely related to the outcome of patients with ICH and may be a biological marker for outcome prediction.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Jing Chen
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Wen-Yao Zhu
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Ting Zhao
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Qi Zhong
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Kai Zhou
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Zhao-You Meng
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Yan-Chun Wang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Peng-Fei Wang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Huang Fang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
867
|
Yang Y, Zhang M, Kang X, Jiang C, Zhang H, Wang P, Li J. Impaired adult hippocampal neurogenesis and cognitive ability in a mouse model of intrastriatal hemorrhage. Neurosci Lett 2015; 599:133-9. [PMID: 26021875 DOI: 10.1016/j.neulet.2015.05.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022]
Abstract
Thrombin released by hematoma is an important mediator of the secondary injury of intracerebral hemorrhage (ICH), however, the effect of thrombin on adult neurogenesis and cognitive ability remains elusive. In this study, intrastriatal injection of 0.05 U thrombin didn't affect the neurogenesis at the subgranular zone (SGZ), which was distal to the injection site. 0.1 U thrombin increased the 5-bromo-2-deoxyuridine(+) (BrdU(+), S-phase proliferating cells)/doublecortin(+) (DCX(+), immature neurons) double labelled neurons, but decreased BrdU(+)/NeuN(+) double labelled mature neurons. Higher doses of thrombin (1 U, 2 U, and 5 U) significantly decreased the BrdU(+)/DCX(+) and BrdU(+)/NeuN(+) double labelled cells. After 1 U thrombin injection, cell apoptosis was found at the dentate gyrus of hippocampus at 3-24 h, but not 5 d post-injury. Thrombin infusion (1 U) induced spatial memory deficits in Morris water maze test; whereas, hirudin, the thrombin antagonist, significantly reversed both neurogenesis loss and spatial learning and memory impairment. In conclusion, at least at short term (5 days) after striatum ICH, the effect of high dose of thrombin on neurogenesis of SGZ, and the spatial learning and memory ability, is detrimental.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Neurological, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Meikui Zhang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China.
| | - Xiaoni Kang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Chen Jiang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Huan Zhang
- Department Students Brigade, The Second Military Medical University, No. 800, Xiangyin Road, Shanghai 200433, China
| | - Pei Wang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Jingjing Li
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| |
Collapse
|
868
|
Pan W, Yan Q, Qin M, Jin G, Sun J, Ning X, Zhuang W, Peng B, Li G. Detection of cerebral hemorrhage in rabbits by time-difference magnetic inductive phase shift spectroscopy. PLoS One 2015; 10:e0128127. [PMID: 26001112 PMCID: PMC4441421 DOI: 10.1371/journal.pone.0128127] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 04/22/2015] [Indexed: 11/18/2022] Open
Abstract
Cerebral hemorrhage, a difficult issue in clinical practice, is often detected and studied with computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). However, these expensive devices are not readily available in economically underdeveloped regions, and hence are unable to provide bedside and emergency on-site monitoring. The magnetic inductive phase shift (MIPS) is an emerging technology that may become a new tool to detect cerebral hemorrhage and to serve as an inexpensive partial substitute to medical imaging. In order to study a wider band of cerebral hemorrhage MIPS and to provide more useful information for measuring cerebral hemorrhage, we established a cerebral hemorrhage magnetic induction phase shift spectroscopy (MIPSS) detection system. Thirteen rabbits with five cerebral hemorrhage states were studied using a single coil-coil within a 1 MHz-200 MHz frequency range in linear sweep. A feature band (FB) with the highest detection sensitivity and the greatest stability was selected for further analysis and processing. In addition, a maximum conductivity cerebrospinal fluid (CSF) MRI was performed to verify and interpret the MIPSS result. The average phase shift change induced by a 3 ml injection of autologous blood under FB was -7.7503° ± 1.4204°, which was considerably larger than our previous work. Data analysis with a non-parametric statistical Friedman M test showed that in the FB, MIPSS could distinguish the five states of cerebral hemorrhage in rabbits, with a statistical significance of p<0.05. A B-F distribution profile was designed according to the MIPSS under FB that can provide instantaneous diagnostic information about the cerebral hemorrhage severity from a single set of measurements. The results illustrate that the MIPSS detection method is able to provide a new possibility for real-time monitoring and diagnosis of the severity of cerebral hemorrhage.
Collapse
Affiliation(s)
- Wencai Pan
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Qingguang Yan
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Surgery Institute of the Third Military Medical University, Chongqing, China
| | - Mingxin Qin
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Gui Jin
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Jian Sun
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Xu Ning
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Wei Zhuang
- College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Bin Peng
- Department of Biomedical Engineering, Chongqing University, Chongqing, China
| | - Gen Li
- Department of Biomedical Engineering, Chongqing University, Chongqing, China
| |
Collapse
|
869
|
Hackett MJ, DeSouza M, Caine S, Bewer B, Nichol H, Paterson PG, Colbourne F. A new method to image heme-Fe, total Fe, and aggregated protein levels after intracerebral hemorrhage. ACS Chem Neurosci 2015; 6:761-70. [PMID: 25695130 DOI: 10.1021/acschemneuro.5b00037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An intracerebral hemorrhage (ICH) is a devastating stroke that results in high mortality and significant disability in survivors. Unfortunately, the underlying mechanisms of this injury are not yet fully understood. After the primary (mechanical) trauma, secondary degenerative events contribute to ongoing cell death in the peri-hematoma region. Oxidative stress is thought to be a key reason for this delayed injury, which is likely due to free-Fe-catalyzed free radical reactions. Unfortunately, this is difficult to prove with conventional biochemical assays that fail to differentiate between alterations that occur within the hematoma and peri-hematoma zone. This is a critical limitation, as the hematoma contains tissue severely damaged by the initial hemorrhage and is unsalvageable, whereas the peri-hematoma region is less damaged but at risk from secondary degenerative events. Such events include oxidative stress mediated by free Fe presumed to originate from hemoglobin breakdown. Therefore, minimizing the damage caused by oxidative stress following hemoglobin breakdown and Fe release is a major therapeutic target. However, the extent to which free Fe contributes to the pathogenesis of ICH remains unknown. This investigation used a novel imaging approach that employed resonance Raman spectroscopic mapping of hemoglobin, X-ray fluorescence microscopic mapping of total Fe, and Fourier transform infrared spectroscopic imaging of aggregated protein following ICH in rats. This multimodal spectroscopic approach was used to accurately define the hematoma/peri-hematoma boundary and quantify the Fe concentration and the relative aggregated protein content, as a marker of oxidative stress, within each region. The results revealed total Fe is substantially increased in the hematoma (0.90 μg cm(-2)), and a subtle but significant increase in Fe that is not in the chemical form of hemoglobin is present within the peri-hematoma zone (0.32 μg cm(-2)) within 1 day of ICH, relative to sham animals (0.22 μg cm(-2)). Levels of aggregated protein were significantly increased within both the hematoma (integrated band area 0.10 AU) and peri-hematoma zone (integrated band area 0.10 AU) relative to sham animals (integrated band area 0.056 AU), but no significant difference in aggregated protein content was observed between the hematoma and peri-hematoma zone. This result suggests that the chemical form of Fe and its ability to generate free radicals is likely to be a more critical predictor of tissue damage than the total Fe content of the tissue. Furthermore, this article describes a novel approach to colocalize nonheme Fe and aggregated protein in the peri-hematoma zone following ICH, a significant methodological advancement for the field.
Collapse
Affiliation(s)
- Mark J. Hackett
- Molecular
and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Mauren DeSouza
- Department
of Psychology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
- Stress,
Memory and Behaviour Lab, Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana, Rio Grande do Sul 97500-970, Brazil
| | - Sally Caine
- Department
of Anatomy and Cell Biology, University of Saskatchewan, 107
Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Brian Bewer
- Canadian Light Source, Saskatoon, Saskatchewan S7N 2V3, Canada
| | - Helen Nichol
- Department
of Anatomy and Cell Biology, University of Saskatchewan, 107
Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Phyllis G. Paterson
- College of
Pharmacy and Nutrition, University of Saskatchewan, D Wing Health Sciences, 107 Wiggins
Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Frederick Colbourne
- Department
of Psychology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| |
Collapse
|
870
|
Zhao X, Sun G, Zhang J, Ting SM, Gonzales N, Aronowski J. Dimethyl Fumarate Protects Brain From Damage Produced by Intracerebral Hemorrhage by Mechanism Involving Nrf2. Stroke 2015; 46:1923-8. [PMID: 25977275 DOI: 10.1161/strokeaha.115.009398] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/23/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE Intracerebral hemorrhage (ICH) represents a devastating form of stroke for which there is no effective treatment. This preclinical study was designed to evaluate dimethyl fumarate (DMF), a substance recently approved for the treatment of multiple sclerosis, as therapy for ICH. We hypothesized that DMF through activating the master regulator of cellular self-defense responses, transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), would act as effective treatment for ICH-mediated damage. METHODS Male rats and mice, including Nrf2 knockouts, were subjected to intracerebral injection of blood (to mimic ICH) and then treated with DMF. Neurological deficit, brain edema, gene induction profile and hematoma resolution were evaluated. Phagocytic functions of primary microglia in culture were used to study hematoma resolution. RESULTS Treatment with DMF induced Nrf2-target genes, improved hematoma resolution, reduced brain edema, and ultimately enhanced neurological recovery in rats and wild-type, but not Nrf2 knockout, mice. Most importantly, the treatment of ICH with DMF showed a 24 h window of therapeutic opportunity. CONCLUSIONS A clinically relevant dose of DMF demonstrates potent therapeutic efficacy and impressive 24 h therapeutic window of opportunity. This study merits further evaluation of this compound as potential treatment for ICH in humans.
Collapse
Affiliation(s)
- Xiurong Zhao
- From the Stroke Program-Department of Neurology, University of Texas-Houston, Medical School
| | - Guanghua Sun
- From the Stroke Program-Department of Neurology, University of Texas-Houston, Medical School
| | - Jie Zhang
- From the Stroke Program-Department of Neurology, University of Texas-Houston, Medical School
| | - Shun-Ming Ting
- From the Stroke Program-Department of Neurology, University of Texas-Houston, Medical School
| | - Nicole Gonzales
- From the Stroke Program-Department of Neurology, University of Texas-Houston, Medical School
| | - Jaroslaw Aronowski
- From the Stroke Program-Department of Neurology, University of Texas-Houston, Medical School.
| |
Collapse
|
871
|
Guo X, Li H, Zhang Z, Li S, Zhang L, Zhang J, Han G. Hyperglycemia and Mortality Risk in Patients with Primary Intracerebral Hemorrhage: A Meta-Analysis. Mol Neurobiol 2015; 53:2269-75. [PMID: 25972238 DOI: 10.1007/s12035-015-9184-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/21/2015] [Indexed: 11/30/2022]
Abstract
Hyperglycemia may be associated with worse functional outcomes in patients with primary intracerebral hemorrhage. We performed a systematic review and meta-analysis to investigate the relationship between hyperglycemia and mortality risk in patients with primary intracerebral hemorrhage. We searched PubMed and Embase databases for studies investigating the association between hyperglycemia and mortality risk in patients with primary intracerebral hemorrhage. We estimated the pooled relative risk (RR) with its 95% confidence interval (95% CI) to assess the impact of hyperglycemia on mortality risk. Seventeen studies with a total of 6527 primary intracerebral hemorrhage patients were included. Meta-analysis of those studies showed that hyperglycemia significantly increased risk of mortality in patients with primary intracerebral hemorrhage (RR = 2.36, 95% CI 1.79-3.12). Subgroup analysis by time of follow-up showed that hyperglycemia significantly increased risk of short-term mortality (RR = 3.97, 95% CI 2.13-7.43) and long-term mortality (RR = 1.53, 95% CI 1.14-2.05). The RR of mortality for per 1-mmol/L increment in glucose level was 1.14 (95% CI 1.06-1.22). In patients with primary intracerebral hemorrhage, hyperglycemia significantly increases risk of both short-term mortality and long-term mortality.
Collapse
Affiliation(s)
- Xiaoming Guo
- Neurosurgery Department, The First Affiliated Hospital of General Hospital of PLA, Beijing, 100048, China.
| | - Helin Li
- Neurology Department, The First Affiliated Hospital of General Hospital of PLA, Beijing, 100048, China
| | - Zhiwen Zhang
- Neurosurgery Department, The First Affiliated Hospital of General Hospital of PLA, Beijing, 100048, China
| | - Shouchun Li
- Neurosurgery Department, The First Affiliated Hospital of General Hospital of PLA, Beijing, 100048, China
| | - Lizhi Zhang
- Neurosurgery Department, The First Affiliated Hospital of General Hospital of PLA, Beijing, 100048, China
| | - Jiajing Zhang
- Neurosurgery Department, Air Force General Hospital of PLA, Beijing, 100048, China
| | - Guiqing Han
- Department of Medicine, Guizhou Hospital, Guiyang, 550002, China
| |
Collapse
|
872
|
Stem cell-based therapies for intracerebral hemorrhage in animal model: a meta-analysis. Neurol Sci 2015; 36:1311-7. [PMID: 25972140 DOI: 10.1007/s10072-015-2238-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/09/2015] [Indexed: 11/27/2022]
Abstract
Stem cell to be a new intervention for treating intracerebral hemorrhage (ICH) might benefit humans. Therefore, we collected animal studies to find the effect of this innovative treatment. In July 2014, we searched Medline (from 1950), Embase (from 1980), China Biology Medicine disk (from 1978) for studies on stem cells used for treating experimental ICH in animal models that reported neurobehavioral and structural outcome. We evaluated the quality of these studies and used a weighted mean difference random affects model for the meta-analysis. We have collected 30 studies from 650 publications identified through systematic review describing the effects of 5 different type of stem cells on 12 different neurobehavioral scales with 1101 rodents or monkeys. Although there is lack of uniformity of the evaluation methods, these researches showed consistent improvements both in neurobehavioral function and structural outcomes. Besides, the quality of these studies needs to be raised. In conclusion, stem cells hold extensive potential in treating ICH, which should be further evaluated with more evidence-based, high-quality animal studies.
Collapse
|
873
|
Fingolimod alters inflammatory mediators and vascular permeability in intracerebral hemorrhage. Neurosci Bull 2015; 31:755-62. [PMID: 25958190 DOI: 10.1007/s12264-015-1532-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023] Open
Abstract
Intracerebral hemorrhage (ICH) leads to high rates of death and disability. The pronounced inflammatory reactions that rapidly follow ICH contribute to disease progression. Our recent clinical trial demonstrated that oral administration of an immune modulator fingolimod restrained secondary injury derived from initial hematoma, but the mechanisms remain unknown. In this study, we aim to investigate the effects of fingolimod on inflammatory mediators and vascular permeability in the clinical trial of oral fingolimod for intracerebral hemorrhage (ICH). The results showed that fingolimod decreased the numbers of circulating CD4(+) T, CD8(+) T, CD19(+) B, NK, and NKT cells and they recovered quickly after the drug was stopped. The plasma ICAM level was decreased and IL-10 was increased by fingolimod. Interestingly, fingolimod protected vascular permeability as indicated by a decreased plasma level of MMP9 and the reduced rT1%. In conclusion, modulation of systemic inflammation by fingolimod demonstrates that it is an effective therapeutic agent for ICH. Fingolimod may prevent perihematomal edema enlargement by protecting vascular permeability.
Collapse
|
874
|
Wang C, Fei Y, Xu C, Zhao Y, Pan Y. Bone marrow mesenchymal stem cells ameliorate neurological deficits and blood-brain barrier dysfunction after intracerebral hemorrhage in spontaneously hypertensive rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:4715-4724. [PMID: 26191161 PMCID: PMC4503033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/14/2015] [Indexed: 06/04/2023]
Abstract
Spontaneous intracerebral hemorrhage (ICH) is a common and fatal subtype of stroke, with hypertension the most common cause of this disorder. Bone marrow derived mesenchymal stem cells (BM-MSCs) have been shown to elicit protective properties in stroke models. In the present study, male spontaneously hypertensive rats (SHR) were subjected to ICH by intracerebral injection with autologous blood, Wistar-Kyoto (WKY) rats were employed as control. The neurological function outcomes and blood-brain barrier (BBB) were assessed after BM-MSCs transplantation. Our results showed that BM-MSCs grafts via the tail vein significantly decreased the modified neurological severity score (mNSS) and the modified limb placing test (MLPT) score at 14 days after ICH, and the scores were gradually lowered till the end of test. Furthermore, BM-MSCs transplantation effectively attenuated the BBB permeability compared with the vehicle only group, as evidenced by the low level of Evans blue leakage in the BM-MSC group. In addition, we found that BM-MSCs grafts elevated the levels of tight junction associated protein occludin, and type IV collagen. Taken together, our results suggest that intravenously transplanted BM-MSCs exert therapeutic effects on ICH in spontaneously hypertensive rats. The underlying mechanisms are associated with the enhanced neurological function recovery and increased integrity of BBB. Our results provide the increased understanding of the underlying mechanisms and perspective of BMSCs in treatment for stroke.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, People’s Republic of China
| | - Yiping Fei
- Hematologic Cancer Center, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, People’s Republic of China
| | - Congshu Xu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, People’s Republic of China
| | - Yue Zhao
- Department of Neurology, Harbin Second HospitalHarbin 150026, People’s Republic of China
| | - Yujun Pan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, People’s Republic of China
| |
Collapse
|
875
|
Toxic role of prostaglandin E2 receptor EP1 after intracerebral hemorrhage in mice. Brain Behav Immun 2015; 46:293-310. [PMID: 25697396 PMCID: PMC4422065 DOI: 10.1016/j.bbi.2015.02.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/28/2015] [Accepted: 02/08/2015] [Indexed: 02/07/2023] Open
Abstract
Inflammatory mechanisms mediated by prostaglandins may contribute to the progression of intracerebral hemorrhage (ICH)-induced brain injury, but they are not fully understood. In this study, we examined the effect of prostaglandin E2 receptor EP1 (EP1R) activation and inhibition on brain injury in mouse models of ICH and investigated the underlying mechanism of action. ICH was induced by injecting collagenase, autologous blood, or thrombin into the striatum of middle-aged male and female mice and aged male mice. Effects of selective EP1R agonist ONO-DI-004, antagonist SC51089, and nonspecific Src family kinase inhibitor PP2 were evaluated by a combination of histologic, magnetic resonance imaging (MRI), immunofluorescence, molecular, cellular, and behavioral assessments. EP1R was expressed primarily in neurons and axons but not in astrocytes or microglia after ICH induced by collagenase. In middle-aged male mice subjected to collagenase-induced ICH, EP1R inhibition mitigated brain injury, brain edema, cell death, neuronal degeneration, neuroinflammation, and neurobehavioral deficits, whereas its activation exacerbated these outcomes. EP1R inhibition also was protective in middle-aged female mice and aged male mice after collagenase-induced ICH and in middle-aged male mice after blood- or thrombin-induced ICH. EP1R inhibition also reduced oxidative stress, white matter injury, and brain atrophy and improved functional outcomes. Histologic results were confirmed by MRI. Src kinase phosphorylation and matrix metalloproteinase-9 activity were increased by EP1R activation and decreased by EP1R inhibition. EP1R regulated matrix metalloproteinase-9 activity through Src kinase signaling, which mediated EP1R toxicity after collagenase-induced ICH. We conclude that prostaglandin E2 EP1R activation plays a toxic role after ICH through mechanisms that involve the Src kinases and the matrix metalloproteinase-9 signaling pathway. EP1R inhibition could be a novel therapeutic strategy to improve outcomes after ICH.
Collapse
|
876
|
Abstract
Stroke is a leading cause of adult disability in the United States. However, limited number of molecularly targeted therapy exists for stroke. Recent studies have shown that Li-pocalin-2 (LCN2) is an acute phase protein mediating neuroinflammation after ischemic and hemorrhagic strokes. This review is an attempt to summarize some LCN2-related research findings and discuss its role in stroke.
Collapse
Affiliation(s)
- Wen-Hai Chou
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44224, USA
| | - Guona Wang
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44224, USA
| | - Varun Kumar
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44224, USA
| | - Yi-Chinn Weng
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44224, USA
| |
Collapse
|
877
|
Egashira Y, Hua Y, Keep RF, Xi G. Intercellular cross-talk in intracerebral hemorrhage. Brain Res 2015; 1623:97-109. [PMID: 25863131 DOI: 10.1016/j.brainres.2015.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disorder with high mortality and morbidity. Currently, there are few treatment strategies for ICH-induced brain injury. A recent increase in interest in the pathophysiology of ICH has led to elucidation of the pathways underlying ICH-induced brain injury, pathways where intercellular and hematoma to cell signaling play important roles. In this review, we summarize recent advances in ICH research focusing on intercellular and hematoma:cell cross-talk related to brain injury and recovery after ICH. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yusuke Egashira
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
878
|
Yang J, Arima H, Wu G, Heeley E, Delcourt C, Zhou J, Chen G, Wang X, Zhang S, Yu S, Chalmers J, Anderson CS. Prognostic Significance of Perihematomal Edema in Acute Intracerebral Hemorrhage. Stroke 2015; 46:1009-13. [PMID: 25712944 DOI: 10.1161/strokeaha.114.007154] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jie Yang
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Hisatomi Arima
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Guojun Wu
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Emma Heeley
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Candice Delcourt
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Junshan Zhou
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Guofang Chen
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Xia Wang
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Shihong Zhang
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Sungwook Yu
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - John Chalmers
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| | - Craig S. Anderson
- From the Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China (J.Y., J.Z.); The George Institute for Global Health, Central Clinical School, University of Sydney and Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (J.Y., H.A., G.W., E.H., C.D., G.C., X.W., S.Z., S.Y., J.C., C.S.A.); Department of Neurology, Hebei Yutian Hospital, Tangshan, China (G.W.); Department of Neurology, Xuzhou Central Hospital, Xuzhou, China (G
| |
Collapse
|
879
|
Abstract
Microglia are considered the brain's resident immune cell involved in immune defense, immunocompetence, and phagocytosis. They maintain tissue homeostasis within the brain and spinal cord under normal condition and serves as its initial host defense system. However, when the central nervous system (CNS) faces injury, microglia respond through signaling molecules expressed or released by neighboring cells. Microglial responses are dual in nature. They induce a nonspecific immune response that may exacerbate CNS injury, especially in the acute stages, but are also essential to CNS recovery and repair. The full range of microglial mechanisms have yet to be clarified, but there is accumulating knowledge about microglial activation in acute CNS injury. Microglial responses require hours to days to fully develop, and may present a therapeutic target for intervention with a much longer window of opportunity compare to other neurological treatments. The challenge will be to find ways to selectively suppress the deleterious effects of microglial activation without compromising its beneficial functions. This review aims to provide an overview of the recent progress relating on the deleterious and beneficial effect of microglia in the setting of acute CNS injury and the potential therapeutic intervention against microglial activation to CNS injury.
Collapse
Affiliation(s)
- Masahito Kawabori
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA, 94121, USA
| | | |
Collapse
|
880
|
Ke K, Song Y, Shen J, Niu M, Zhang H, Yuan D, Ni H, Zhang Y, Liu X, Dai A, Cao M. Up-regulation of Glis2 involves in neuronal apoptosis after intracerebral hemorrhage in adult rats. Cell Mol Neurobiol 2015; 35:345-354. [PMID: 25370802 PMCID: PMC11486247 DOI: 10.1007/s10571-014-0130-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/24/2014] [Indexed: 12/26/2022]
Abstract
The novel Krüppel-like zinc finger protein Gli-similar 2 (Glis2), one member of the transcription factors, is involved in controlling the flow of genetic information and the modulation of diverse cellular activities. Accumulating evidence has demonstrated its important roles in adult development and several diseases. However, information regarding the regulation and possible function of Glis2 in the central nervous system is still limited. In this study, we explored the roles of Glis2 during the pathophysiological process of intracerebral hemorrhage (ICH). An ICH rat model was established and assessed by behavioral tests. Expression of Glis2 was significantly up-regulated in brain areas surrounding the hematoma following ICH. Immunofluorescence showed that Glis2 was strikingly increased in neurons, but not astrocytes or microglia. Up-regulation of Glis2 was found to be accompanied by the increased expression of active caspase-3 and Bax and decreased expression of Bcl-2 in vivo and vitro studies. Moreover, knocking down Glis2 by RNA-interference in PC12 cells reduced active caspase-3 and Bax expression while increased Bcl-2. Collectively, we speculated that Glis2 might exert pro-apoptotic function in neurons following ICH.
Collapse
Affiliation(s)
- Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Yan Song
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Mu Niu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Haiyan Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Daming Yuan
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Haidan Ni
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Yu Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Xiaorong Liu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Aihua Dai
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
881
|
Kathirvelu B, Carmichael ST. Intracerebral hemorrhage in mouse models: therapeutic interventions and functional recovery. Metab Brain Dis 2015; 30:449-59. [PMID: 24810632 PMCID: PMC4226812 DOI: 10.1007/s11011-014-9559-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 04/28/2014] [Indexed: 12/11/2022]
Abstract
There has been strong pre-clinical research on mechanisms of initial cell death and tissue injury in intracerebral hemorrhage (ICH). This data has led to the evaluation of several therapeutics for neuroprotection or the mitigation of early tissue damage. Most of these studies have been done in the rat. Also, there has been little study of the mechanisms of tissue repair and recovery. This review examines the testing of candidate therapeutics in mouse models of ICH for their effect on tissue protection and repair. This review will help the readers compare it to the extensively researched rat model of ICH and thus enhance work that are pending in mouse model.
Collapse
Affiliation(s)
- Balachandar Kathirvelu
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA,
| | | |
Collapse
|
882
|
Sun J, Wei ZZ, Gu X, Zhang JY, Zhang Y, Li J, Wei L. Intranasal delivery of hypoxia-preconditioned bone marrow-derived mesenchymal stem cells enhanced regenerative effects after intracerebral hemorrhagic stroke in mice. Exp Neurol 2015; 272:78-87. [PMID: 25797577 DOI: 10.1016/j.expneurol.2015.03.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/11/2022]
Abstract
Intracerebral hemorrhagic stroke (ICH) causes high mortality and morbidity with very limited treatment options. Cell-based therapy has emerged as a novel approach to replace damaged brain tissues and promote regenerative processes. In this study we tested the hypothesis that intranasally delivered hypoxia-preconditioned BMSCs could reach the brain, promote tissue repair and improve functional recovery after ICH. Hemorrhagic stroke was induced in adult C57/B6 mice by injection of collagenase IV into the striatum. Animals were randomly divided into three groups: sham group, intranasal BMSC treatment group, and vehicle treatment group. BMSCs were pre-treated with hypoxic preconditioning (HP) and pre-labeled with Hoechst before transplantation. Behavior tests, including the mNSS score, rotarod test, adhesive removal test, and locomotor function evaluation were performed at varying days, up to 21days, after ICH to evaluate the therapeutic effects of BMSC transplantation. Western blots and immunohistochemistry were performed to analyze the neurotrophic effects. Intranasally delivered HP-BMSCs were identified in peri-injury regions. NeuN+/BrdU+ co-labeled cells were markedly increased around the hematoma region, and growth factors, including BDNF, GDNF, and VEGF were significantly upregulated in the ICH brain after BMSC treatment. The BMSC treatment group showed significant improvement in behavioral performance compared with the vehicle group. Our data also showed that intranasally delivered HP-BMSCs migrated to peri-injury regions and provided growth factors to increase neurogenesis after ICH. We conclude that intranasal administration of BMSC is an effective treatment for ICH, and that it enhanced neuroregenerative effects and promoted neurological functional recovery after ICH. Overall, the investigation supports the potential therapeutic strategy for BMSC transplantation therapy against hemorrhagic stroke.
Collapse
Affiliation(s)
- Jinmei Sun
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Laboratories of Stem Cell Biology and Neural Regeneration and Function Recovery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zheng Zachory Wei
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Laboratories of Stem Cell Biology and Neural Regeneration and Function Recovery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James Ya Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Laboratories of Stem Cell Biology and Neural Regeneration and Function Recovery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jimei Li
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Laboratories of Stem Cell Biology and Neural Regeneration and Function Recovery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| | - Ling Wei
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Laboratories of Stem Cell Biology and Neural Regeneration and Function Recovery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
883
|
Wu G, Wu J, Jiao Y, Wang L, Wang F, Zhang Y. Rosiglitazone infusion therapy following minimally invasive surgery for intracerebral hemorrhage evacuation decreases matrix metalloproteinase-9 and blood-brain barrier disruption in rabbits. BMC Neurol 2015; 15:37. [PMID: 26021445 PMCID: PMC4472168 DOI: 10.1186/s12883-015-0287-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 02/25/2015] [Indexed: 01/10/2023] Open
Abstract
Background The objective of this study was to investigate the effects of Rosiglitazone (RSG) infusion therapy following minimally invasive surgery (MIS) for intracerebral hemorrhage(ICH) evacuation on perihematomal secondary brain damage as assessed by MMP-9 levels, blood–brain barrier (BBB) permeability and neurological function. Methods A total of 40 male rabbits (2.8–3.4 kg) was randomly assigned to a normal control group (NC group; 10 rabbits), a model control group (MC group; 10 rabbits), a minimally invasive treatment group (MIS group; 10 rabbits) or a combined MIS and RSG group (MIS + RSG group; 10 rabbits). ICH was induced in all the animals, except for the NC group. MIS was performed to evacuate ICH 6 hours after the successful preparation of the ICH model in the MIS and MIS + RSG groups. The animals in the MC group underwent the same procedures for ICH evacuation but without hematoma aspiration, and the NC group was subjected to sham surgical procedures. The neurological deficit scores (Purdy score) and ICH volumes were determined on days 1, 3 and 7. All of the animals were sacrificed on day 7, and the perihematomal brain tissue was removed to determine the levels of PPARγ, MMP-9, BBB permeability and brain water content (BWC). Results The Purdy score, perihematomal PPARγ levels, BBB permeability, and BWC were all significantly increased in the MC group compared to the NC group. After performing the MIS for evacuating the ICH, the Purdy score and the ICH volume were decreased on days 1, 3 and 7 compared to the MC group. A remarkable decrease in perihematomal levels of PPARγ, MMP-9, BBB permeability and BWC were observed. The MIS + RSG group displayed a remarkable increase in PPARγ as well as significant decrease in MMP-9, BBB permeability and BWC compared with the MIS group. Conclusions RSG infusion therapy following MIS for ICH treatment might be more efficacious for reducing the levels of MMP-9 and secondary brain damage than MIS therapy alone.
Collapse
Affiliation(s)
- Guofeng Wu
- Emergency Department, Guizhou Medical University, No. 28, Guiyijie Road, Liuguangmen, Postal code 550004, Guiyang City, Guizhou Province, People's Republic of China.
| | - Junjie Wu
- Department of Neurology of Affiliated Hospital, Guizhou Medical University, No. 28, Guiyijie Road, Liuguangmen, Postal code 550004, Guiyang City, Guizhou Province, People's Republic of China.
| | - Yu Jiao
- Department of Neurology of Affiliated Hospital, Guizhou Medical University, No. 28, Guiyijie Road, Liuguangmen, Postal code 550004, Guiyang City, Guizhou Province, People's Republic of China.
| | - Likun Wang
- Emergency Department, Guizhou Medical University, No. 28, Guiyijie Road, Liuguangmen, Postal code 550004, Guiyang City, Guizhou Province, People's Republic of China.
| | - Fan Wang
- Department of Neurology of Affiliated Hospital, Guizhou Medical University, No. 28, Guiyijie Road, Liuguangmen, Postal code 550004, Guiyang City, Guizhou Province, People's Republic of China.
| | - Yingjun Zhang
- Department of Medical Images of Affiliated Hospital, Guizhou Medical University, No. 28, Guiyijie Road, Liuguangmen, Postal code 550004, Guiyang City, Guizhou Province, People's Republic of China.
| |
Collapse
|
884
|
Ni W, Okauchi M, Hatakeyama T, Gu Y, Keep RF, Xi G, Hua Y. Deferoxamine reduces intracerebral hemorrhage-induced white matter damage in aged rats. Exp Neurol 2015; 272:128-34. [PMID: 25749188 DOI: 10.1016/j.expneurol.2015.02.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/23/2015] [Accepted: 02/27/2015] [Indexed: 12/11/2022]
Abstract
Iron contributes to c-Jun N-terminal kinases (JNK) activation in young rats and white matter injury in piglets after intracerebral hemorrhage (ICH). In the present study, we examined the effect of deferoxamine on ICH-induced white matter injury and JNK activation and in aged rats. Male Fischer 344 rats (18months old) had either an intracaudate injection of 100μl of autologous blood or a needle insertion (sham). The rats were treated with deferoxamine or vehicle with different regimen (dosage, duration and time window). White matter injury and activation of JNK were examined. We found that a dose of DFX should be at more than 10mg/kg for a therapeutic duration more than 2days with a therapeutic time window of 12h to reduce ICH-induced white matter loss at 2months. ICH-induced white matter injury was associated with JNK activation. The protein levels of phosphorylated-JNK (P-JNK) were upregulated at day-1 after ICH and then gradually decreased. P-JNK immunoreactivity was mostly located in white matter bundles. ICH-induced JNK activation was reduced by DFX treatment. This study demonstrated that DFX can reduce ICH-induced JNK activation and white matter damage.
Collapse
Affiliation(s)
- Wei Ni
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Masanobu Okauchi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Yuxiang Gu
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
885
|
Urday S, Beslow LA, Goldstein DW, Vashkevich A, Ayres AM, Battey TWK, Selim MH, Kimberly WT, Rosand J, Sheth KN. Measurement of perihematomal edema in intracerebral hemorrhage. Stroke 2015; 46:1116-9. [PMID: 25721012 DOI: 10.1161/strokeaha.114.007565] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Perihematomal edema (PHE) is a marker of secondary injury in intracerebral hemorrhage (ICH). PHE measurement on computed tomography (CT) is challenging, and the principles used to detect PHE have not been described fully. We developed a systematic approach for CT-based measurement of PHE. METHODS Two independent raters measured PHE volumes on baseline and 24-hour post-ICH CT scans of 20 primary supratentorial ICH subjects. Boundaries were outlined with an edge-detection tool and adjusted after inspection of the 3 orthogonal planes. PHE was delineated with the additional principle that it should be (a) more hypodense than the corresponding area in the contralateral hemisphere and (b) most hypodense immediately surrounding the hemorrhage. We examined intra- and interrater reliability using intraclass correlation coefficients and Bland-Altman plots for interrater consistency. CT-based PHE was also compared using magnetic resonance imaging-based PHE detection for 18 subjects. RESULTS Median PHE volumes were 22.7 cc at baseline and 20.4 cc at 24 hours post-ICH. There were no statistically significant differences in PHE measurements between raters. Interrater and intrarater reliability for PHE were excellent. At baseline and 24 hours, interrater intraclass correlation coefficients were 0.98 (0.96-1.00) and 0.98 (0.97-1.00); intrarater intraclass correlation coefficients were 0.99 (0.99-1.00) and 0.99 (0.98-1.00). Bland-Altman analysis showed the bias for PHE measurements at baseline and 24 hours, -0.5 cc (SD, 5.4) and -3.2 cc (SD, 5.0), was acceptably small. PHE volumes determined by CT and magnetic resonance imaging were similar (23.9±16.9 cc versus 23.9±16.0 cc, R(2) = 0.98, P<0.0001). CONCLUSIONS Our method measures PHE with excellent reliability at baseline and 24 hours post-ICH.
Collapse
Affiliation(s)
- Sebastian Urday
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Lauren A Beslow
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - David W Goldstein
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Anastasia Vashkevich
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Alison M Ayres
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Thomas W K Battey
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Magdy H Selim
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - W Taylor Kimberly
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Jonathan Rosand
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.)
| | - Kevin N Sheth
- From the Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT (S.U., L.A.B., D.W.G., K.N.S.); Center for Human Genetic Research, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (A.V., A.M.A., T.W.K.B., W.T.K., J.R.); and Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.H.S.).
| |
Collapse
|
886
|
Shi L, Qin J, Song B, Wang QM, Zhang R, Liu X, Liu Y, Hou H, Chen X, Ma X, Jiang C, Sun X, Gong G, Xu Y. Increased frequency of circulating regulatory T cells in patients with acute cerebral hemorrhage. Neurosci Lett 2015; 591:115-120. [PMID: 25703227 DOI: 10.1016/j.neulet.2015.02.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/01/2015] [Accepted: 02/17/2015] [Indexed: 01/09/2023]
Abstract
Cerebral hemorrhage (ICH) is a serious stroke subtype, currently lacking effective treatment. Recent research has shown that CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) play a key role in the immune response of ischemic stroke. However, Tregs in human hemorrhagic stroke are poorly investigated. In this study, a total of 90 ICH patients and 60 healthy controls were recruited. The frequency of circulating Tregs, plasma levels of TGF-β and IL-10, and the severity of neural dysfunction in ICH patients were investigated at different time points post ICH. We found that the peripheral frequency of Tregs in ICH patients was significantly increased, accompanied by boosted activated T cells. Importantly, the elevation of circulating Tregs in patients with severe dysfunction was much higher than that in less-severe patients, suggesting that disease severity affects circulating Tregs to exert regulatory function. Furthermore, both TGF-β and IL-10 that are related to the function of Tregs, were also increased in the peripheral blood of ICH patients. Our results demonstrate that Tregs-mediated immune imbalance might affect the development and severity of ICH, and suggest that Tregs may be used as tools and targets of cellular immunotherapy to effectively treat acute hemorrhagic stroke.
Collapse
Affiliation(s)
- Lijin Shi
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China; Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Henan Province 453100, China
| | - Jie Qin
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Bo Song
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Qing Mei Wang
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Rui Zhang
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Xinjing Liu
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Yutao Liu
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Haiman Hou
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Xiulan Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinxiang Medical University, Henan Province 453100, China
| | - Xun Ma
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Chenyang Jiang
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Xiao Sun
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Guangming Gong
- Department of Microbiology and Immunology, Basic Medical College of Zhengzhou University, Henan Province 450000, China.
| | - Yuming Xu
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China.
| |
Collapse
|
887
|
Abstract
Intraventricular hemorrhage (IVH) is a severity factor and treatment target in intracerebral hemorrhage. This study aimed to investigate whether systemic edaravone, a free-radical scavenger, could attenuate the brain injury after IVH in a rat model. Our findings showed that an intraventricular injection of autologous whole blood resulted in acute brain edema, increased malondialdehyde level, and decreased superoxide dismutase enzyme activity. Immediate edaravone treatment after IVH can reduce IVH-induced brain edema and elevated lipid peroxidation. Furthermore, repeated edaravone treatment (immediately, 24 h, and 48 h after IVH) improved the IVH-induced learning and memory damage. These effects suggest that edaravone may be a potential therapeutic agent for IVH, especially those intracerebral hemorrhage patients with ventricular extension.
Collapse
|
888
|
Zheng M, Du H, Ni W, Koch LG, Britton SL, Keep RF, Xi G, Hua Y. Iron-induced necrotic brain cell death in rats with different aerobic capacity. Transl Stroke Res 2015; 6:215-23. [PMID: 25649272 DOI: 10.1007/s12975-015-0388-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/16/2015] [Accepted: 01/21/2015] [Indexed: 12/29/2022]
Abstract
Brain iron overload has a key role in brain injury after intracerebral hemorrhage (ICH). Our recent study demonstrated that ICH-induced brain injury was greater in low capacity runner (LCR) than in high capacity runner (HCR) rats. The present study examines whether iron-induced brain injury differs between LCRs and HCRs. Adult male LCR and HCR rats had an intracaudate injection of iron or saline. Rats were euthanized at 2 and at 24 h after T2 magnetic resonance imaging, and the brains were used for immunostaining and Western blotting. LCRs had more hemispheric swelling, T2 lesion volumes, blood-brain barrier disruption, and neuronal death at 24 h after iron injection (p < 0.05). Many propidium iodide (PI)-positive cells, indicative of necrotic cell death, were observed in the ipsilateral basal ganglia of both HCRs and LCRs at 2 h after iron injection. PI fluorescence intensity was higher in LCRs than in HCRs. In addition, membrane attack complex (MAC) expression was increased at 2 h after iron injection and was higher in LCRs than in HCRs. The PI-positive cells co-localized with MAC-positive cells in the ipsilateral basal ganglia. Iron induces more severe necrotic brain cell death, brain swelling, and blood-brain barrier disruption in LCR rats, which may be related with complement activation and MAC formation.
Collapse
Affiliation(s)
- Mingzhe Zheng
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | | | | | | | | | | | | | | |
Collapse
|
889
|
Urday S, Kimberly WT, Beslow LA, Vortmeyer AO, Selim MH, Rosand J, Simard JM, Sheth KN. Targeting secondary injury in intracerebral haemorrhage--perihaematomal oedema. Nat Rev Neurol 2015; 11:111-22. [PMID: 25623787 DOI: 10.1038/nrneurol.2014.264] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Perihaematomal oedema (PHO) is an important pathophysiological marker of secondary injury in intracerebral haemorrhage (ICH). In this Review, we describe a novel method to conceptualize PHO formation within the framework of Starling's principle of movement of fluid across a capillary wall. We consider progression of PHO through three stages, characterized by ionic oedema (stage 1) and progressive vasogenic oedema (stages 2 and 3). In this context, possible modifiers of PHO volume and their value in identifying patients who would benefit from therapies that target secondary injury are discussed; the practicalities of using neuroimaging to measure PHO volume are also considered. We examine whether PHO can be used as a predictor of neurological outcome following ICH, and we provide an overview of emerging therapies. Our discussion emphasizes that PHO has clinical relevance both as a therapeutic target, owing to its augmentation of the mass effect of a haemorrhage, and as a surrogate marker for novel interventions that target secondary injury.
Collapse
Affiliation(s)
- Sebastian Urday
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - W Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Lauren A Beslow
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Alexander O Vortmeyer
- Department of Pathology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Magdy H Selim
- Department of Neurology, Beth Israel Deaconess Medical Centre, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Kevin N Sheth
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| |
Collapse
|
890
|
Su X, Wang H, Kang D, Zhu J, Sun Q, Li T, Ding K. Necrostatin-1 ameliorates intracerebral hemorrhage-induced brain injury in mice through inhibiting RIP1/RIP3 pathway. Neurochem Res 2015; 40:643-50. [PMID: 25576092 DOI: 10.1007/s11064-014-1510-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/20/2014] [Accepted: 12/26/2014] [Indexed: 12/16/2022]
Abstract
Necroptosis is a recently discovered programmed necrosis, regulated by receptor interacting protein kinase 1 (RIP1) and RIP3 after death signal stimulation and could be specifically inhibited by necrostatin-1. The aim of this study was to investigate the role of RIP1 and RIP3 signal pathways in a mouse model of collagenase-induced intracerebral hemorrhage (ICH) and assess the effect of necrostatin-1 on brain injury after ICH. We found that RIP1 and RIP3 proteins were abundantly expressed and increased in mice brain after ICH. Necrostatin-1 pretreatment improved neurological function and attenuated brain edema in mice after ICH. Moreover, necrostatin-1 reduced RIP1-RIP3 interaction and propidium iodide (PI) positive cell death, and further inhibited microglia activation and pro-inflammatory mediator genes [tumor necrosis factor-a (TNF-α) and interleukin-1β (IL-1β)] expression after ICH. These findings indicate that RIP1/RIP3-mediated necroptosis is an important mechanism of cell death after ICH. Through inhibiting necroptosis, necrostatin-1 plays a protective role in reducing necrotic cell death after ICH. Necrostatin-1 is a promising therapeutic agent that protects cells from necroptosis and improves functional outcome.
Collapse
Affiliation(s)
- Xingfen Su
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu Province, People's Republic of China,
| | | | | | | | | | | | | |
Collapse
|
891
|
Guan J, Zhang B, Zhang J, Ding W, Xiao Z, Zhu Z, Han Q, Wu C, Sun Y, Tong W, Dai J, Wang R. Nerve regeneration and functional recovery by collagen-binding brain-derived neurotrophic factor in an intracerebral hemorrhage model. Tissue Eng Part A 2015; 21:62-74. [PMID: 24941993 DOI: 10.1089/ten.tea.2014.0139] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) exerts therapeutic effects following intracerebral hemorrhage (ICH). However, it is difficult to maintain sufficient concentrations in the hemorrhage hemisphere. We demonstrated previously that BDNF fused to a collagen-binding domain (CBD) could bind to collagen in the ventricular ependyma and stimulate cell proliferation in the subventricular zone (SVZ). In this study, we verified the therapeutic effects of CBD-BDNF in the rat ICH model induced by bacterial collagenase by injecting CBD-BDNF into the lateral ventricle of ICH rats. The results demonstrated that CBD-BDNF was retained at high levels in the hemorrhage hemisphere, where it promoted neural regeneration and angiogenesis, reduced tissue loss, and improved functional recovery.
Collapse
Affiliation(s)
- Jian Guan
- 1 Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
892
|
Jiao X, He P, Li Y, Fan Z, Si M, Xie Q, Chang X, Huang D. The Role of Circulating Tight Junction Proteins in Evaluating Blood Brain Barrier Disruption following Intracranial Hemorrhage. DISEASE MARKERS 2015; 2015:860120. [PMID: 26586924 PMCID: PMC4637473 DOI: 10.1155/2015/860120] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/14/2015] [Accepted: 09/16/2015] [Indexed: 02/05/2023]
Abstract
Brain injury after intracranial hemorrhage (ICH) results in significant morbidity and mortality. Blood brain barrier (BBB) disruption is a hallmark of ICH-induced brain injury; however, data mirroring BBB disruption in human ICH are scarce. The aim of this study was to assess the significance of circulating biomarkers in evaluating BBB disruption after ICH. Twenty-two patients with ICH were recruited in this study. Concentrations of the tight junction proteins (TJs) Claudin-5 (CLDN5), Occludin (OCLN), and zonula occludens 1 (ZO-1) and vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) were measured by using enzyme-linked immunosorbent assay in serum and cerebrospinal fluid (CSF) samples obtained from patients with ICH. The white blood cell (WBC) count in blood and CSF, albumin (ALB) levels in the CSF (ALBCSF), and the BBB ratio were significantly higher in the ICH than in controls (p < 0.05). Significantly higher levels of CLDN5, OCLN, ZO-1, MMP-9, and VEGF in CSF were observed in the ICH group; these biomarkers were also positively associated with BBB ratio (p < 0.05). Our data revealed that circulating TJs could be considered the potential biomarkers reflecting the integrity of the BBB in ICH.
Collapse
Affiliation(s)
- Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Ping He
- Shantou University Medical College, Guangdong 515041, China
| | - Yazhen Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Zhicheng Fan
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Mengya Si
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
- *Dongyang Huang:
| |
Collapse
|
893
|
Abstract
Intracranial hemorrhage (ICH) affects 0.2-0.5 % of atrial fibrillation (AF) patients taking a novel oral anticoagulant (NOAC) each year. About two thirds of ICHs are intracerebral and one quarter subdural. The 30-day case fatality of NOAC-associated ICH was similar to that of warfarin-associated ICH in two trials. Consistent predictors of ICH are increasing age, a history of prior stroke or TIA, and concomitant use of an antiplatelet drug. Compared to warfarin, the NOACs significantly reduce the risk of ICH by half (risk ratio = 0.44; 95 % CI: 0.37 to 0.51). Compared to aspirin, apixaban has a similar risk of ICH (risk ratio = 0.84; 95 % CI, 0.38 to 1.87). Current treatments for NOAC-associated ICH include nonactivated and activated prothrombin complex concentrate, which reverse the anticoagulant effects of the NOACs, but their effects on bleeding and patient outcome are not known. Future treatments for NOAC-associated ICH promise to include specific antidotes to dabigatran (e.g., aDabi-Fab, PER977) and factor Xa inhibitors (e.g., r-Antidote PRT064445, PER977).
Collapse
|
894
|
Zhang Y, Yi B, Ma J, Zhang L, Zhang H, Yang Y, Dai Y. Quercetin promotes neuronal and behavioral recovery by suppressing inflammatory response and apoptosis in a rat model of intracerebral hemorrhage. Neurochem Res 2014; 40:195-203. [PMID: 25543848 DOI: 10.1007/s11064-014-1457-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/18/2014] [Accepted: 10/16/2014] [Indexed: 10/24/2022]
Abstract
Intracerebral hemorrhage (ICH) is a common and devastating disease affecting millions of people worldwide annually. Exaggerated inflammation and apoptosis are two pivotal pathological processes for secondary brain injury after ICH. Quercetin, a flavonoid widely distributed in various herbs, fruits and vegetables, has been proved to improve neuronal functional recovery in spinal cord injury rats. However, the efficacy of quercetin in caring for post-ICH brain injury has not been investigated. In the present study, we established an ICH model by injecting type VII bacterial collagenase (0.5U) into the central striatum of male Sprague-Dawley rats. The animals were randomized to four groups: sham-operation group; ICH + vehicle group; ICH + 5 mg/kg quercetin group; and ICH + 50 mg/kg quercetin group. The expression levels of IL-1β, IL-4, IL-6 and TNF-α in the brain tissue were assayed by Real-time PCR, ELISA and Western Blot, and cell apoptosis was assayed by TUNEL and caspase-3 staining 3 days after model establishment. It was found that the lesion volume, the brain water content, the expression levels of the four inflammation markers and the number of apoptotic cells were reduced significantly in ICH rats receiving quercetin, especially in 50 mg/kg quercetin group. These results confirmed the therapeutic efficacy of quercetin in repairing brain injury, probably by inhibiting inflammatory response and apoptosis, thus promoting nerve functional restoration.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
895
|
Chen S, Yang Q, Chen G, Zhang JH. An Update on Inflammation in the Acute Phase of Intracerebral Hemorrhage. Transl Stroke Res 2014; 6:4-8. [DOI: 10.1007/s12975-014-0384-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022]
|
896
|
Kothandaraman A, Anson T, Reynolds A. Effect of low voltage AC fields on cardiovascular implants. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 46:497-503. [PMID: 25492014 DOI: 10.1016/j.msec.2014.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/03/2014] [Accepted: 10/19/2014] [Indexed: 11/16/2022]
Abstract
Coronary Artery Stents have been the preferred form of treatment for vascular occlusive disease, due to the minimally invasive surgical procedure, post-operative recovery time and cost, when compared to open coronary bypass surgery. The cellular response upon applying an AC electric field to type 316LM Stainless Steel stent mimics was investigated in this paper. The highest RBC adhesion was observed at voltages higher than 88 mV and lower than 74 mV. Their unique alignment along the lines of fracture on the stent surface at 88 mV was a phenomenon caused by an increase in electrical conductivity in these regions. Being able to control RBC adhesion may have various clinical implications such as inhibition of thrombus formation, and provide a basis to analyse whether electric fields may be applied to cancer therapy as well.
Collapse
Affiliation(s)
| | - Tony Anson
- Brunel University, Kingston Lane, Uxbridge, Middlesex UB8 3PH, UK
| | - Alan Reynolds
- Brunel University, Kingston Lane, Uxbridge, Middlesex UB8 3PH, UK
| |
Collapse
|
897
|
Taylor RA, Hammond MD, Ai Y, Sansing LH. CX3CR1 signaling on monocytes is dispensable after intracerebral hemorrhage. PLoS One 2014; 9:e114472. [PMID: 25469644 PMCID: PMC4255025 DOI: 10.1371/journal.pone.0114472] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/07/2014] [Indexed: 12/20/2022] Open
Abstract
Intracerebral hemorrhage is a subset of stroke for which there is no specific treatment. The Ly6Chi CCR2+ monocytes have been shown to contribute to acute injury after intracerebral hemorrhage. The other murine monocyte subset expresses CX3CR1 and lower Ly6C levels, and contributes to repair in other disease models. We hypothesized that the Ly6Clo CX3CR1+ monocytes would contribute to recovery after intracerebral hemorrhage. Intracerebral hemorrhage was modeled by blood injection in WT and CX3CR1-null bone marrow chimeras. Neurological outcomes and leukocyte recruitment were quantified at various time points. Functional outcomes were equal at 1, 3, 7, and 14 days after intracerebral hemorrhage in both genotypes. No differences were observed in leukocyte recruitment between genotypes on either 3 or 7 days after intracerebral hemorrhage. A few hundred Ly6Clo monocytes were found in the ipsilateral hemisphere in each genotype and they did not change over time. Peripherally derived CX3CR1+ monocytes were observed in the perihematomal brain 7 and 14 days after intracerebral hemorrhage. Our data suggests CX3CR1 signaling on monocytes does not play an influential role in acute injury or functional recovery after intracerebral hemorrhage and therefore CX3CR1 is not a therapeutic target to improve outcome after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Roslyn A. Taylor
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Matthew D. Hammond
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Youxi Ai
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
898
|
Senn R, Elkind MSV, Montaner J, Christ-Crain M, Katan M. Potential role of blood biomarkers in the management of nontraumatic intracerebral hemorrhage. Cerebrovasc Dis 2014; 38:395-409. [PMID: 25471997 DOI: 10.1159/000366470] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/06/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH), a subtype of stroke associated with high mortality and disability, accounts for 13% of all strokes. Basic and clinical research has contributed to our understanding of the complex pathophysiology of neuronal injury in ICH. Outcome rates, however, remain stable, and questions regarding acute management of ICH remain unanswered. Newer research is aiming at matching measured levels of serum proteins, enzymes, or cells to different stages of brain damage, suggesting that blood biomarkers may assist in acute diagnosis, therapeutic decisions, and prognostication. This paper provides an overview on the most promising blood biomarkers and their potential role in the diagnosis and management of spontaneous ICH. SUMMARY Information was collected from studies, reviews, and guidelines listed in PubMed up to November 2013 on blood biomarkers of nontraumatic ICH in humans. We describe the potential role and limitations of GFAP, S100B/RAGE, and ApoC-III as diagnostic biomarkers, β-Amyloid as a biomarker for etiological classification, and 27 biomarkers for prognosis of mortality and functional outcome. Within the group of prognostic markers we discuss markers involved in coagulation processes (e.g., D-Dimers), neuroendocrine markers (e.g., copeptin), systemic metabolic markers (e.g., blood glucose levels), markers of inflammation (e.g., IL-6), as well as growth factors (e.g., VEGF), and others (e.g., glutamate). Some of those blood biomarkers are agents of pathologic processes associated with hemorrhagic stroke but also other diseases, whereas others play more distinct pathophysiological roles and help in understanding the basic mechanisms of brain damage and/or recovery in ICH. KEY MESSAGES Numerous blood biomarkers are associated with different pathophysiological pathways in ICH, and some of them promise to be useful in the management of ICH, eventually contributing additional information to current tools for diagnosis, therapy monitoring, risk stratification, or intervention. Up to date, however, no blood biomarker of ICH has been studied sufficiently to find its way into clinical routine yet; well-designed, large-scale, clinical studies addressing relevant clinical questions are needed. We suggest that the effectiveness of biomarker research in ICH might be improved by international cooperation and shared resources for large validation studies, such as provided by the consortium on stroke biomarker research (http://stroke-biomarkers.com/page.php?title=Resources).
Collapse
Affiliation(s)
- Rebecca Senn
- Department of Endocrinology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
899
|
Shen J, Liu Y, Song Y, Li L, Duan C, Zhou Y, Ke K. CHMP4B, ESCRT-III associating protein, associated with neuronal apoptosis following intracerebral hemorrhage. Brain Res 2014; 1597:1-13. [PMID: 25478783 DOI: 10.1016/j.brainres.2014.11.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 12/17/2022]
Abstract
Charged multivesicular body protein (CHMP) represents a family of small helical proteins that contain an N-terminal basically charged region and a smaller C-terminal acidic region, which are highly conserved in all eukaryotes. CHMP4B, a core component of the endosomal sorting complex required for transport (ESCRT)-III, is requisite for endosomal sorting and other biological processes. Here, we demonstrate that CHMP4B may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). From the results of Western blot, immunohistochemistry and immunofluorescence, we obtained a significant up-regulation of CHMP4B in neurons adjacent to the hematoma following ICH. Increasing CHMP4B level was found to be accompanied by the up-regulation of Fas receptor (Fas), Fas ligand (FasL), active caspase-8, and active caspase-3. Besides, CHMP4B co-localized well with Fas and active caspase-3 in neurons, indicating its potential role in neuronal apoptosis. What's more, our in vitro study, using CHMP4B RNA interference in PC12 cells, further confirmed that CHMP4B might exert its pro-apoptotic function on neuronal apoptosis through extrinsic pathway. Thus, CHMP4B may play a role in promoting the brain damage following ICH.
Collapse
Affiliation(s)
- Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yonghua Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Song
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Lei Li
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Chengwei Duan
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ying Zhou
- Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
900
|
Wowk S, Ma Y, Colbourne F. Mild Therapeutic Hypothermia Does Not Reduce Thrombin-Induced Brain Injury. Ther Hypothermia Temp Manag 2014; 4:180-7. [DOI: 10.1089/ther.2014.0014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shannon Wowk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Yonglie Ma
- Department of Psychology, University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Psychology, University of Alberta, Edmonton, Canada
| |
Collapse
|