1
|
Keuters MH, Antila S, Immonen R, Plotnikova L, Wojciechowski S, Lehtonen S, Alitalo K, Koistinaho J, Dhungana H. The Impact of VEGF-C-Induced Dural Lymphatic Vessel Growth on Ischemic Stroke Pathology. Transl Stroke Res 2024:10.1007/s12975-024-01262-9. [PMID: 38822994 DOI: 10.1007/s12975-024-01262-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/15/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024]
Abstract
Timely relief of edema and clearance of waste products, as well as promotion of anti-inflammatory immune responses, reduce ischemic stroke pathology, and attenuate harmful long-term effects post-stroke. The discovery of an extensive and functional lymphatic vessel system in the outermost meningeal layer, dura mater, has opened up new possibilities to facilitate post-stroke recovery by inducing dural lymphatic vessel (dLV) growth via a single injection of a vector encoding vascular endothelial growth factor C (VEGF-C). In the present study, we aimed to improve post-stroke outcomes by inducing dLV growth in mice. We injected mice with a single intracerebroventricular dose of adeno-associated viral particles encoding VEGF-C before subjecting them to transient middle cerebral artery occlusion (tMCAo). Behavioral testing, Gadolinium (Gd) contrast agent-enhanced magnetic resonance imaging (MRI), and immunohistochemical analysis were performed to define the impact of VEGF-C on the post-stroke outcome. VEGF-C improved stroke-induced behavioral deficits, such as gait disturbances and neurological deficits, ameliorated post-stroke inflammation, and enhanced an alternative glial immune response. Importantly, VEGF-C treatment increased the drainage of brain interstitial fluid (ISF) and cerebrospinal fluid (CSF), as shown by Gd-enhanced MRI. These outcomes were closely associated with an increase in the growth of dLVs around the region where we observed increased vefgc mRNA expression within the brain, including the olfactory bulb, cortex, and cerebellum. Strikingly, VEGF-C-treated ischemic mice exhibited a faster and stronger Gd-signal accumulation in ischemic core area and an enhanced fluid outflow via the cribriform plate. In conclusion, the VEGF-C-induced dLV growth improved the overall outcome post-stroke, indicating that VEGF-C has potential to be included in the treatment strategies of post-ischemic stroke. However, to maximize the therapeutic potential of VEGF-C treatment, further studies on the impact of an enhanced dural lymphatic system at clinically relevant time points are essential.
Collapse
Affiliation(s)
- Meike Hedwig Keuters
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014, Helsinki, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Salli Antila
- Wihuri Research Institute and Translational Cancer Medicine Program, University of Helsinki, 00014, Helsinki, Finland
| | - Riikka Immonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Lidiia Plotnikova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Sara Wojciechowski
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Sarka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, University of Helsinki, 00014, Helsinki, Finland
| | - Jari Koistinaho
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014, Helsinki, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, 00014, Helsinki, Finland
| | - Hiramani Dhungana
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014, Helsinki, Finland.
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland.
| |
Collapse
|
2
|
Jiang Z, Xu D, Li H, Wu X. A Novel Nomogram to Predict Symptomatic Intracranial Hemorrhage in Ischemic Stroke Patients After Intravenous Thrombolysis. Ther Clin Risk Manag 2023; 19:993-1003. [PMID: 38050618 PMCID: PMC10693780 DOI: 10.2147/tcrm.s436145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/12/2023] [Indexed: 12/06/2023] Open
Abstract
Objective This study aimed to create and validate a novel nomogram to predict the risk of symptomatic intracranial hemorrhage (sICH) in patients with acute ischemic stroke (AIS) who underwent intravenous thrombolysis (IVT). Methods In this retrospective study, 784 patients with AIS who received IVT were enrolled. The patients were randomly divided into two groups: a training set (n=550, 70%) and a testing set (n=234, 30%). Utilizing multivariable logistic regression analysis, relevant factors for the predictive nomogram were selected. The performance of the nomogram was evaluated using various metrics, including the area under the receiver operating characteristic curve (AUC-ROC), the Hosmer-Lemeshow goodness-of-fit test, calibration plots, and decision curve analysis (DCA). Results Multivariable logistic regression analysis showed that specific factors, including National Institutes of Health Stroke Scale (NIHSS) scores, Early infarct signs (EIS), and serum sodium, were identified as independent predictors of sICH. Subsequently, a nomogram was constructed using these predictors. The AUC-ROC values of the nomogram were 0.864 (95% CI: 0.810-0.919) and 0.831 (95% CI: 0.770-0.891) in the training and the validation sets, respectively. Both the calibration plots and the Hosmer-Lemeshow goodness-of-fit test showed favorable agreement in both the training and the validation sets. Additionally, the DCA indicated the practical clinical utility of the nomogram. Conclusion The novel nomogram, which included NIHSS, EIS and serum sodium as variables, had the potential for predicting the risk of sICH in patients with AIS after IVT.
Collapse
Affiliation(s)
- Zhuangzhuang Jiang
- Department of Neurology, Dongyang People’s Hospital, Affiliated to Wenzhou Medical University, Dongyang, Zhejiang, People’s Republic of China
| | - Dongjuan Xu
- Department of Neurology, Dongyang People’s Hospital, Affiliated to Wenzhou Medical University, Dongyang, Zhejiang, People’s Republic of China
| | - Hongfei Li
- Department of Neurology, Dongyang People’s Hospital, Affiliated to Wenzhou Medical University, Dongyang, Zhejiang, People’s Republic of China
| | - Xiaolan Wu
- Department of Neurology, Dongyang People’s Hospital, Affiliated to Wenzhou Medical University, Dongyang, Zhejiang, People’s Republic of China
| |
Collapse
|
3
|
Kung TFC, Wilkinson CM, Liddle LJ, Colbourne F. A systematic review and meta-analysis on the efficacy of glibenclamide in animal models of intracerebral hemorrhage. PLoS One 2023; 18:e0292033. [PMID: 37756302 PMCID: PMC10529582 DOI: 10.1371/journal.pone.0292033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating stroke with many mechanisms of injury. Edema worsens outcome and can lead to mortality after ICH. Glibenclamide (GLC), a sulfonylurea 1- transient receptor potential melastatin 4 (Sur1-Trpm4) channel blocker, has been shown to attenuate edema in ischemic stroke models, raising the possibility of benefit in ICH. This meta-analysis synthesizes current pre-clinical (rodent) literature regarding the efficacy of post-ICH GLC administration (vs. vehicle controls) on behaviour (i.e., neurological deficit, motor, and memory outcomes), edema, hematoma volume, and injury volume. Six studies (5 in rats and 1 in mice) were included in our meta-analysis (PROSPERO registration = CRD42021283614). GLC significantly improved behaviour (standardized mean difference (SMD) = -0.63, [-1.16, -0.09], n = 70-74) and reduced edema (SMD = -0.91, [-1.64, -0.18], n = 70), but did not affect hematoma volume (SMD = 0.0788, [-0.5631, 0.7207], n = 18-20), or injury volume (SMD = 0.2892, [-0.4950, 1.0734], n = 24). However, these results should be interpreted cautiously. Findings were conflicted with 2 negative and 4 positive reports, and Egger regressions indicated missing negative edema data (p = 0.0001), and possible missing negative behavioural data (p = 0.0766). Experimental quality assessed via the SYRCLE and CAMARADES checklists was concerning, as most studies demonstrated high risks of bias. Studies were generally low-powered (e.g., average n = 14.4 for behaviour), and future studies should employ sample sizes of 41 to detect our observed effect size in behaviour and 33 to detect our observed effect in edema. Overall, missing negative studies, low study quality, high risk of bias, and incomplete attention to key recommendations (e.g., investigating female, aged, and co-morbid animals) suggest that further high-powered confirmatory studies are needed before conclusive statements about GLC's efficacy in ICH can be made, and before further clinical trials are performed.
Collapse
Affiliation(s)
- Tiffany F. C. Kung
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Lane J. Liddle
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
Richard S, Gabriel S, John S, Emmanuel M, John-Mary V. The focused quantitative EEG bio-marker in studying childhood atrophic encephalopathy. Sci Rep 2022; 12:13437. [PMID: 35927445 PMCID: PMC9352776 DOI: 10.1038/s41598-022-17062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 07/20/2022] [Indexed: 11/12/2022] Open
Abstract
Although it is a normal involution process in advanced age, brain atrophy—also termed atrophic encephalopathy—can also occur prematurely in childhood as a consequential effect of brain tissues injury through trauma or central nervous system infection, though in both normal and premature occurrences this condition always presents with loss of volume relative to the skull. A common tool for the functional study of brain activities is an electroencephalogram, but analyses of this have reportedly identified mismatches between qualitative and quantitative forms, particularly in the use of Delta-alpha ratio (DAR) indices, meaning that the values may be case dependent. The current study thus examines the value of Focused Occipital Beta-Alpha Ratio (FOBAR) as a modified biomarker for evaluating brain functional changes resulting from brain atrophy. This cross-sectional design study involves 260 patients under 18 years of age. Specifically, 207 patients with brain atrophy are compared with 53 control subjects with CT scan-proven normal brain volume. All the children underwent digital electroencephalography with brain mapping. Results show that alpha posterior dominant rhythm was present in 88 atrophic children and 44 controls. Beta as posterior dominant rhythm was present in an overwhelming 91.5% of atrophic subjects, with 0.009 p-values. The focused occipital Beta-alpha ratio correlated significantly with brain volume loss presented in diagonal brain fraction. The FOBAR and DAR values of the QEEG showed no significant correlation. This work concludes that QEEG cerebral dysfunctional studies may be etiologically and case dependent from the nature of the brain injury. Also, the focused Beta-alpha ratio of the QEEG is a prospective and potential biomarker of consideration in studying childhood atrophic encephalopathy.
Collapse
Affiliation(s)
- Sungura Richard
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela-African Institution of Science and Technology, Arusha, Tanzania.
| | - Shirima Gabriel
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela-African Institution of Science and Technology, Arusha, Tanzania
| | - Spitsbergen John
- Department of Neuroscience, Western Michigan University, Kalamazoo, MI, USA
| | - Mpolya Emmanuel
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela-African Institution of Science and Technology, Arusha, Tanzania
| | - Vianney John-Mary
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela-African Institution of Science and Technology, Arusha, Tanzania
| |
Collapse
|
5
|
He L, Guo ZN, Qu Y, Jin H. Hyponatremia Is Associated With Post-thrombolysis Hemorrhagic Transformation and Poor Clinical Outcome in Ischemic Stroke Patients. Front Mol Neurosci 2022; 15:879863. [PMID: 35923753 PMCID: PMC9341483 DOI: 10.3389/fnmol.2022.879863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveHyponatremia is the most common electrolyte disorder encountered in patients with neurological conditions, such as stroke. Studies have shown that it is associated with worse clinical outcomes and increased mortality in acute ischemic stroke (AIS). However, the role of hyponatremia has not been elucidated in patients with AIS who received intravenous thrombolysis (IVT) therapy. Therefore, this study aimed to investigate the effect of serum sodium levels on the clinical outcome and hemorrhagic transformation (HT) in patients with AIS who received thrombolytic therapy.MethodsPatients diagnosed with AIS who received IVT therapy between May 2015 and December 2020 were included in this study. All patients were screened for serum sodium levels immediately after hospital admission, before IVT therapy. The occurrence of HT was evaluated using computed tomography (CT) 24 ± 2 h after thrombolysis. Then, 3-month clinical outcomes were obtained by telephone calls or outpatient visits, and poor 3-month clinical outcomes were defined as modified Rankin Scale scores ≥3. The effects of serum sodium levels on the clinical outcome and HT were assessed using the multivariate logistic regression analysis.ResultsOf the 963 included patients, 82 (8.5%) had hyponatremia, 157 (16.3%) developed HT, and 333 (34.6%) had poor 3-month outcomes. Of the 82 patients with hyponatremia, 21 (25.6%) developed HT, and 39 (47.6%) had poor 3-month outcomes. Patients with hyponatremia had a higher incidence of post-thrombolysis HT (25.6 vs. 15.4%, p = 0.017) and worse clinical outcome (47.6 vs. 33.4%, p = 0.01) than those with normal serum sodium levels. Patients had significantly lower serum sodium levels in those with HT [138.4 (136.4–140.3, IQR) vs. 139.0 (137.2–140.7, IQR) mmol/L, p = 0.019] and poor 3 month outcome [139.0 (137.2–140.7) vs. 138.4 (136.7–140.3) mmol/L, p = 0.005] than those without. After adjusting for major covariates, the multivariate logistic regression analysis revealed that lower serum sodium levels were independently associated with an increased risk of HT [odds ratio (OR) = 1.804; 95% CI: 1.048–3.105] and poor 3-month outcome (OR = 1.647; 95% CI: 1.012–2.679).ConclusionLower serum sodium level was an independent risk factor for post-thrombolysis HT and poor clinical outcome in patients with AIS who received thrombolytic therapy.
Collapse
Affiliation(s)
- Ling He
- Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Zhen-Ni Guo
- Stroke Center, The First Hospital of Jilin University, Changchun, China
- Department of Neurology, Stroke Center, Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yang Qu
- Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Stroke Center, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Hang Jin,
| |
Collapse
|
6
|
Guse K, Hagemann N, Thiele L, Remlinger J, Salmen A, Hoepner R, Keller I, Meyer P, Grandgirard D, Leib SL, Vassella E, Locatelli G, Hermann DM, Chan A. CNS Antigen-Specific Neuroinflammation Attenuates Ischemic Stroke With Involvement of Polarized Myeloid Cells. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2022; 9:9/4/e1168. [PMID: 35676093 PMCID: PMC9177141 DOI: 10.1212/nxi.0000000000001168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 02/25/2022] [Indexed: 11/15/2022]
Abstract
Background and Objectives Experimental studies indicate shared molecular pathomechanisms in cerebral hypoxia-ischemia and autoimmune neuroinflammation. This has led to clinical studies investigating the effects of immunomodulatory therapies approved in multiple sclerosis on inflammatory damage in stroke. So far, mutual and combined interactions of autoimmune, CNS antigen-specific inflammatory reactions and cerebral ischemia have not been investigated so far. Methods Active MOG35-55 experimental autoimmune encephalomyelitis (EAE) was induced in male C57Bl/6J mice. During different phases of EAE, transient middle cerebral artery occlusion (tMCAO, 60 minutes) was induced. Brain tissue was analyzed for infarct size and immune cell infiltration. Multiplex gene expression analysis was performed for 186 genes associated with neuroinflammation and hypoxic-ischemic damage. Results Mice with severe EAE disease showed a substantial reduction in infarct size after tMCAO. Histopathologic analysis showed less infiltration of CD45+ hematopoietic cells in the infarct core of severely diseased acute EAE mice; this was accompanied by an accumulation of Arginase1-positive/Iba1-positive cells. Gene expression analysis indicated an involvement of myeloid cell-driven anti-inflammatory mechanisms in the attenuation of ischemic injury in severely diseased mice exposed to tMCAO in the acute EAE phase. Discussion CNS autoantigen-specific autoimmunity has a protective influence on primary tissue damage after experimental stroke, indicating a very early involvement of CNS antigen-specific, myeloid cell-associated anti-inflammatory immune mechanisms that mitigate ischemic injury in the acute EAE phase.
Collapse
|
7
|
Pushie MJ, Messmer M, Sylvain NJ, Heppner J, Newton JM, Hou H, Hackett MJ, Kelly ME, Peeling L. Multimodal imaging of hemorrhagic transformation biomarkers in an ischemic stroke model. Metallomics 2022; 14:mfac007. [PMID: 35254441 PMCID: PMC9056027 DOI: 10.1093/mtomcs/mfac007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022]
Abstract
Hemorrhagic transformation of ischemic stroke has devastating consequences, with high mortality and poor functional outcomes. Animal models of ischemic stroke also demonstrate the potential for hemorrhagic transformation, which complicates biochemical characterization, treatment studies, and hinders poststroke functional outcomes in affected subjects. The incidence of hemorrhagic transformation of ischemic stroke in animal model research is not commonly reported. The postmortem brain of such cases presents a complex milieu of biomarkers due to the presence of healthy cells, regions of varying degrees of ischemia, dead and dying cells, dysregulated metabolites, and blood components (especially reactive Fe species released from lysed erythrocytes). To improve the characterization of hemorrhage biomarkers on an ischemic stroke background, we have employed a combination of histology, X-ray fluorescence imaging (XFI), and Fourier transform infrared (FTIR) spectroscopic imaging to assess 122 photothrombotic (ischemic) stroke brains. Rapid freezing preserves brain biomarkers in situ and minimizes metabolic artifacts due to postmortem ischemia. Analysis revealed that 25% of the photothrombotic models had clear signs of hemorrhagic transformation. The XFI and FTIR metabolites provided a quantitative method to differentiate key metabolic regions in these models. Across all hemorrhage cases, it was possible to consistently differentiate otherwise healthy tissue from other metabolically distinct regions, including the ischemic infarct, the ischemic penumbra, blood vessels, sites of hemorrhage, and a region surrounding the hemorrhage core that contained elevated lipid oxidation. Chemical speciation of deposited Fe demonstrates the presence of heme-Fe and accumulation of ferritin.
Collapse
Affiliation(s)
- M J Pushie
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - M Messmer
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - N J Sylvain
- Clinical Trial Support Unit, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - J Heppner
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - J M Newton
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - H Hou
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - M J Hackett
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, AUS
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, AUS
| | - M E Kelly
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - L Peeling
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| |
Collapse
|
8
|
Zeng M, Zhou H, He Y, Du H, Yin J, Hou Y, Zhu J, Zhang Y, Shao C, Yang J, Wan H. Danhong injection enhances the therapeutic effect of mannitol on hemispheric ischemic stroke by ameliorating blood-brain barrier disruption. Biomed Pharmacother 2021; 142:112048. [PMID: 34435588 DOI: 10.1016/j.biopha.2021.112048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/05/2021] [Accepted: 08/12/2021] [Indexed: 01/01/2023] Open
Abstract
Mannitol, a representative of hyperosmolar therapy, is indispensable for the treatment of malignant cerebral infarction, but its therapeutic effect is limited by its exacerbation of blood-brain barrier (BBB) disruption. This study was to explore whether Danhong injection (DHI), a standardized product extracted from Salvia miltiorrhiza Bunge and Carthamus tinctorius L., inhibits the destructive effect of mannitol on BBB and thus enhancing the treatment of hemispheric ischemic stroke. SD rats were subjected to pMCAO followed by intravenous bolus injections of mannitol with/without DHI intervention. Neurological deficit score, brain edema, infarct volume at 24 h after MCAO and histopathology, microvascular ultrastructure, immunohistochemistry and immunofluorescence staining of endothelial cell junctions, energy metabolism in the ischemic penumbra were assessed. Intravenous mannitol after MCAO resulted in a decrease in 24 h mortality and cerebral edema, whereas no significant benefit on neurological deficits, infarct volume and microvascular ultrastructure. Moreover, mannitol led to the loss of endothelial integrity, manifested by the decreased expression of occludin, junctional adhesion molecule-1 (JAM-1) and zonula occluden-1 (ZO-1) and the discontinuity of occludin staining around the periphery of endothelial cells. Meanwhile, after mannitol treatment, energy-dependent vimentin and F-actin, ATP content, and ATP5D expression were down-regulated, while MMP2 and MMP9 expression increased in the ischemic penumbra. All the insults after mannitol treatment were attenuated by addition of intravenous DHI. The results suggest DHI as a potential remedy to attenuate mannitol-related BBB disruption, and the potential of DHI to upregulate energy metabolism and inhibit the activity of MMPs is likely attributable to its effects observed.
Collapse
Affiliation(s)
- Miaolin Zeng
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haixia Du
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Junjun Yin
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yongchun Hou
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiaqi Zhu
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yangyang Zhang
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chongyu Shao
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
9
|
Lecordier S, Manrique-Castano D, El Moghrabi Y, ElAli A. Neurovascular Alterations in Vascular Dementia: Emphasis on Risk Factors. Front Aging Neurosci 2021; 13:727590. [PMID: 34566627 PMCID: PMC8461067 DOI: 10.3389/fnagi.2021.727590] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular dementia (VaD) constitutes the second most prevalent cause of dementia in the world after Alzheimer’s disease (AD). VaD regroups heterogeneous neurological conditions in which the decline of cognitive functions, including executive functions, is associated with structural and functional alterations in the cerebral vasculature. Among these cerebrovascular disorders, major stroke, and cerebral small vessel disease (cSVD) constitute the major risk factors for VaD. These conditions alter neurovascular functions leading to blood-brain barrier (BBB) deregulation, neurovascular coupling dysfunction, and inflammation. Accumulation of neurovascular impairments over time underlies the cognitive function decline associated with VaD. Furthermore, several vascular risk factors, such as hypertension, obesity, and diabetes have been shown to exacerbate neurovascular impairments and thus increase VaD prevalence. Importantly, air pollution constitutes an underestimated risk factor that triggers vascular dysfunction via inflammation and oxidative stress. The review summarizes the current knowledge related to the pathological mechanisms linking neurovascular impairments associated with stroke, cSVD, and vascular risk factors with a particular emphasis on air pollution, to VaD etiology and progression. Furthermore, the review discusses the major challenges to fully elucidate the pathobiology of VaD, as well as research directions to outline new therapeutic interventions.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel Manrique-Castano
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Yara El Moghrabi
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
10
|
Zhou W, Zhang Y, Jiao Y, Yin W, Dong H, Xu S, Tang D, Jiang J, Shao J, Wang Z, Yu W. Dexmedetomidine maintains blood-brain barrier integrity by inhibiting Drp1-related endothelial mitochondrial dysfunction in ischemic stroke. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1177-1188. [PMID: 34244711 DOI: 10.1093/abbs/gmab092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Indexed: 12/17/2022] Open
Abstract
Stroke is the second leading cause of death and long-term disability worldwide, which lacks effective treatment. Perioperative stroke is associated with much higher rates of mortality and disability. The neuroprotective role of dexmedetomidine (Dex), a highly selective agonist of alpha2-adrenergic receptor, has been reported in a stroke rat model, and it was found that pretreatment of Dex before stroke could alleviate blood-brain barrier (BBB) breakdown. However, the underlying mechanisms are still unknown. As the brain endothelial cells are the main constituents of BBB and in high demand of energy, mitochondrial function of endothelial cells plays an important role in the maintenance of BBB. Given that dynamin-related protein 1 (Drp1) is a protein mediating mitochondrial fission, with mitochondrial fusion that balances mitochondrial morphology and ensures mitochondria function, the present study was designed to investigate the possible role of Drp1 in endothelial cells involved in the neuroprotective effects of Dex in ischemic stroke. Our results showed that preconditioning with Dex reduced infarction volume, alleviated brain water content and BBB damage, and improved neurological scores in middle cerebral artery occlusion rats. Meanwhile, Dex enhanced cell activity and decreased cell apoptosis in oxygen-glucose deprivation human brain microvascular endothelial cells in vitro. These protective effects of Dex were correlated with the mitochondrial morphology integrality of endothelial cells, mediated by increased phosphorylation of serine 637 in Drp1, and could be reversed by α2-adrenergic receptor antagonist Yohimbine and AMP-activated protein kinase inhibitor Compound C. These findings suggest new molecular pathways involved in the neuroprotective effects of Dex in ischemic stroke. As Dex is routinely used as a sedative drug clinically, our findings provide molecular evidence that it has perioperative neuroprotection from ischemic stroke.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yunchun Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wen Yin
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Haiping Dong
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Saihong Xu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Junli Jiang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jianlin Shao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, No.1 School of Clinical Medicine, Kunming Medical University, Kunming 650011, China
| | - Zhenhong Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
11
|
Baicalein, Baicalin, and Wogonin: Protective Effects against Ischemia-Induced Neurodegeneration in the Brain and Retina. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8377362. [PMID: 34306315 PMCID: PMC8263226 DOI: 10.1155/2021/8377362] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/08/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Ischemia is a common pathological condition present in many neurodegenerative diseases, including ischemic stroke, retinal vascular occlusion, diabetic retinopathy, and glaucoma, threatening the sight and lives of millions of people globally. Ischemia can trigger excessive oxidative stress, inflammation, and vascular dysfunction, leading to the disruption of tissue homeostasis and, ultimately, cell death. Current therapies are very limited and have a narrow time window for effective treatment. Thus, there is an urgent need to develop more effective therapeutic options for ischemia-induced neural injuries. With emerging reports on the pharmacological properties of natural flavonoids, these compounds present potent antioxidative, anti-inflammatory, and antiapoptotic agents for the treatment of ischemic insults. Three major active flavonoids, baicalein, baicalin, and wogonin, have been extracted from Scutellaria baicalensis Georgi (S. baicalensis); all of which are reported to have low cytotoxicity. They have been demonstrated to exert promising pharmacological capabilities in preventing cell and tissue damage. This review focuses on the therapeutic potentials of these flavonoids against ischemia-induced neurotoxicity and damage in the brain and retina. The bioactivity and bioavailability of baicalein, baicalin, and wogonin are also discussed. It is with hope that the therapeutic potential of these flavonoids can be utilized and developed as natural treatments for ischemia-induced injuries of the central nervous system (CNS).
Collapse
|
12
|
Bordoni L, Li B, Kura S, Boas DA, Sakadžić S, Østergaard L, Frische S, Gutiérrez-Jiménez E. Quantification of Capillary Perfusion in an Animal Model of Acute Intracranial Hypertension. J Neurotrauma 2020; 38:446-454. [PMID: 32998634 DOI: 10.1089/neu.2019.6901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Intracranial hypertension (IH) is a common feature of many pathologies, including brain edema. In the brain, the extended network of capillaries ensures blood flow to meet local metabolic demands. Capillary circulation may be severely affected by IH, but no studies have quantified the effect of intracranial pressure (ICP) and cerebral perfusion pressure (CPP) on capillary perfusion during the development of brain edema. We used optical coherence tomography angiography to quantify relative changes of fractional perfused volume (FPV) in cortical capillaries and simultaneously monitored ICP and blood pressure (BP) in anesthetized male C57Bl/6NTac mice during development of brain edema induced by water intoxication (WI) within 30 min. WI induced severe IH and brain herniation. ICP and CPP reached 90.2 mm Hg and 38.4 mm Hg, respectively. FPV was significantly affected already at normal ICP (ICP <15 mm Hg, slope ≈ -1.46, p < 0.001) and, at the onset of IH (ICP = 20-22 mm Hg), FPV was 17.9 ± 13.3% lower than baseline. A decreasing trend was observed until the ICP peak (Δ%FPV = -43.6 ± 19.2%). In the ICP range of 7-42 mm Hg, relative changes in FPV were significantly correlated with ICP, BP, and CPP (p < 0.001), with ICP and CPP being the best predictors. In conclusion, elevated ICP induces a gradual collapse of the cerebral microvasculature, which is initiated before the clinical threshold of IH. In summary, the estimate of capillary perfusion might be essential in patients with IH to assess the state of the brain microcirculation and to improve the availability of oxygen and nutrients to the brain.
Collapse
Affiliation(s)
- Luca Bordoni
- Institute of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Baoqiang Li
- Brain Cognition and Brain Disease Institute, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Sreekanth Kura
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Leif Østergaard
- Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark.,Department of Neuroradiology, Aarhus University Hospital, Aarhus C, Denmark
| | | | | |
Collapse
|
13
|
Fang Y, Shi H, Ren R, Huang L, Okada T, Lenahan C, Gamdzyk M, Travis ZD, Lu Q, Tang L, Huang Y, Zhou K, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide Attenuates Brain Edema by Protecting Blood-Brain Barrier and Glymphatic System After Subarachnoid Hemorrhage in Rats. Neurotherapeutics 2020; 17:1954-1972. [PMID: 32918234 PMCID: PMC7851266 DOI: 10.1007/s13311-020-00925-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Brain edema is a vital contributor to early brain injury after subarachnoid hemorrhage (SAH), which is responsible for prolonged hospitalization and poor outcomes. Pharmacological therapeutic targets on edema formation have been the focus of research for decades. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to participate in neural development and brain injury. Here, we used PACAP knockout CRISPR to demonstrate that endogenous PACAP plays an endogenous neuroprotective role against brain edema formation after SAH in rats. The exogenous PACAP treatment provided both short- and long-term neurological benefits by preserving the function of the blood-brain barrier and glymphatic system after SAH. Pretreatment of inhibitors of PACAP receptors showed that the PACAP-involved anti-edema effect and neuroprotection after SAH was facilitated by the selective PACAP receptor (PAC1). Further administration of adenylyl cyclase (AC) inhibitor and sulfonylurea receptor 1 (SUR1) CRISPR activator suggested that the AC-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) axis participated in PACAP signaling after SAH, which inhibited the expression of edema-related proteins, SUR1 and aquaporin-4 (AQP4), through SUR1 phosphorylation. Thus, PACAP may serve as a potential clinical treatment to alleviate brain edema in patients with SAH.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Hui Shi
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Reng Ren
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
| | - Takeshi Okada
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
| | - Cameron Lenahan
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Marcin Gamdzyk
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Zachary D Travis
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Qin Lu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lihui Tang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Yi Huang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Keren Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Jiping Tang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, California, USA
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China.
| | - John H Zhang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA.
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA.
- Department of Anesthesiology, Loma Linda University, Loma Linda, California, USA.
| |
Collapse
|
14
|
Sarvari S, Moakedi F, Hone E, Simpkins JW, Ren X. Mechanisms in blood-brain barrier opening and metabolism-challenged cerebrovascular ischemia with emphasis on ischemic stroke. Metab Brain Dis 2020; 35:851-868. [PMID: 32297170 PMCID: PMC7988906 DOI: 10.1007/s11011-020-00573-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Stroke is the leading cause of disability among adults as well as the 2nd leading cause of death globally. Ischemic stroke accounts for about 85% of strokes, and currently, tissue plasminogen activator (tPA), whose therapeutic window is limited to up to 4.5 h for the appropriate population, is the only FDA approved drug in practice and medicine. After a stroke, a cascade of pathophysiological events results in the opening of the blood-brain barrier (BBB) through which further complications, disabilities, and mortality are likely to threaten the patient's health. Strikingly, tPA administration in eligible patients might cause hemorrhagic transformation and sustained damage to BBB integrity. One must, therefore, delineate upon stroke onset which cellular and molecular factors mediate BBB permeability as well as what key roles BBB rupture plays in the pathophysiology of stroke. In this review article, given our past findings of mechanisms underlying BBB opening in stroke animal models, we elucidate cellular, subcellular, and molecular factors involved in BBB permeability after ischemic stroke. The contribution of each factor to stroke severity and outcome is further discussed. Determinant factors in BBB permeability and stroke include mitochondria, miRNAs, matrix metalloproteinases (MMPs), immune cells, cytokines, chemokines, and adhesion proteins. Once these factors are interrogated and their roles in the pathophysiology of stroke are determined, novel targets for drug discovery and development can be uncovered in addition to novel therapeutic avenues for human stroke management.
Collapse
Affiliation(s)
- Sajad Sarvari
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Faezeh Moakedi
- Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Emily Hone
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA
| | - James W Simpkins
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Experimental Stroke Core Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Xuefang Ren
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA.
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA.
- Experimental Stroke Core Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
| |
Collapse
|
15
|
Guo H, Yin A, Ma Y, Fan Z, Tao L, Tang W, Ma Y, Hou W, Cai G, Zhuo L, Zhang J, Li Y, Xiong L. Astroglial N-myc downstream-regulated gene 2 protects the brain from cerebral edema induced by stroke. Glia 2020; 69:281-295. [PMID: 32652708 PMCID: PMC7754347 DOI: 10.1002/glia.23888] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/16/2020] [Accepted: 06/26/2020] [Indexed: 01/14/2023]
Abstract
Brain edema is a grave complication of brain ischemia and is the main cause of herniation and death. Although astrocytic swelling is the main contributor to cytotoxic edema, the molecular mechanism involved in this process remains elusive. N‐myc downstream‐regulated gene 2 (NDRG2), a well‐studied tumor suppressor gene, is mainly expressed in astrocytes in mammalian brains. Here, we found that NDRG2 deficiency leads to worsened cerebral edema, imbalanced Na+ transfer, and astrocyte swelling after ischemia. We also found that NDRG2 deletion in astrocytes dramatically changed the expression and distribution of aquaporin‐4 and Na+‐K+‐ATPase β1, which are strongly associated with cell polarity, in the ischemic brain. Brain edema and astrocyte swelling were significantly alleviated by rescuing the expression of astrocytic Na+‐K+‐ATPase β1 in NDRG2‐knockout mouse brains. In addition, the upregulation of astrocytic NDRG2 by lentiviral constructs notably attenuated brain edema, astrocytic swelling, and blood–brain barrier destruction. Our results indicate a particular role of NDRG2 in maintaining astrocytic polarization to facilitate Na+ and water transfer balance and to protect the brain from ischemic edema. These findings provide insight into NDRG2 as a therapeutic target in cerebral edema.
Collapse
Affiliation(s)
- Hang Guo
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China.,Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - Anqi Yin
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China.,Department of Anesthesiology, Jinling Hospital, Nanjing, China
| | - Yulong Ma
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ze Fan
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - Liang Tao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - Wenhong Tang
- Department of Anesthesiology, The 960th Hospital of PLA, Jinan, China
| | - Yaqun Ma
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - Guohong Cai
- Institute of Neuroscience, The Air Force Military Medical University, Xi'an, China
| | - Lixia Zhuo
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Zhang
- Department of Biochemistry and Molecular Biology, The Air Force Military Medical University, Xi'an, China
| | - Yan Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China.,Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Air Force Military Medical University, Xi'an, China.,Department of Anesthesiology & Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Galdamez LA, Brunstetter TJ, Lee AG, Tarver WJ. Origins of Cerebral Edema: Implications for Spaceflight-Associated Neuro-Ocular Syndrome. J Neuroophthalmol 2020; 40:84-91. [DOI: 10.1097/wno.0000000000000852] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Song Y, Wang LB, Bei Y, Qin DX, Ai LY, Ma QZ, Lin PY. Carvacryl acetate, a semisynthetic monoterpenic ester obtained from essential oils, provides neuroprotection against cerebral ischemia reperfusion-induced oxidative stress injury via the Nrf2 signalling pathway. Food Funct 2020; 11:1754-1763. [DOI: 10.1039/c9fo02037c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Carvacryl acetate (CA) is a semisynthetic monoterpenic ester obtained from essential oils, and it exerts an antioxidation effect.
Collapse
Affiliation(s)
- Ying Song
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
| | - Li-Bo Wang
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
| | - Yun Bei
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
- Department of Pharmacy
| | - Dong-Xu Qin
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
| | - Li-Yao Ai
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
| | - Qi-Zhuang Ma
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
| | - Pei-Yao Lin
- Department of Pharmacology
- Zhejiang University of Technology
- Hangzhou
- P.R. China
| |
Collapse
|
18
|
Peng M, Ling X, Song R, Gao X, Liang Z, Fang F, Cang J. Upregulation of GLT-1 via PI3K/Akt Pathway Contributes to Neuroprotection Induced by Dexmedetomidine. Front Neurol 2019; 10:1041. [PMID: 31611842 PMCID: PMC6776610 DOI: 10.3389/fneur.2019.01041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/13/2019] [Indexed: 12/30/2022] Open
Abstract
Perioperative ischemic stroke usually leads to neurological dysfunction caused by neuron death. During the ischemic condition, excitotoxity due to extracellular glutamate accumulation is a main mechanism of neuron damage. The clearance of glutamate mainly depends on glutamate transporter-1 (GLT-1) which is expressed in astrocytes. Dexmedetomidine, an α2 adrenergic receptor agonist, is proved to induce neuroprotection. This study was set out to investigate the glutamate-related mechanism involved in the neuroprotective effect of dexmedetomidine. Middle cerebral artery occlusion (MCAO) was used as a model of ischemic stroke in our study. We determined Neurological deficit scores (NDS) and Magnetic resonance imaging (MRI) at three points (2, 6, and 24 h) after middle cerebral artery occlusion (MCAO) to evaluate the neuroprotective effect of dexmedetomidine. Besides, we performed western blot (6 and 24 h after MACO) and immunofluorescent staining (24 h after MCAO) to observe the expression of GLT-1. The effect and mechanism of dexmedetomidine on GLT-1 in primary cultured astrocytes were investigated using western blot and RT-PCR. Our results showed that pretreatment with dexmedetomidine improved NDS and reduced infarct volume as well as upregulating GLT-1 expression. Furthermore, using Atipamezole and LY294002, we found that dexmedetomidine significantly increased GLT-1 levels in astrocytes via activating α2 adrenergic receptor and PI3K/AKT pathway both in vitro and in vivo study. Overall, our present study indicated that dexmedetomidine had neuroprotective effects on ischemia stroke and upregulation of GLT-1 levels by PI3K/AKT dependent pathway might be the potential mechanism.
Collapse
Affiliation(s)
- Mengyuan Peng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruixue Song
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuan Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhifeng Liang
- Comparative Nerve Imaging Study Group, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fang Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Cang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Shadman J, Sadeghian N, Moradi A, Bohlooli S, Panahpour H. Magnesium sulfate protects blood-brain barrier integrity and reduces brain edema after acute ischemic stroke in rats. Metab Brain Dis 2019; 34:1221-1229. [PMID: 31037556 DOI: 10.1007/s11011-019-00419-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 04/16/2019] [Indexed: 11/27/2022]
Abstract
Brain edema is a fatal complication of acute ischemic stroke and associated with worse outcomes in patients. This study was designed to evaluate the effects of magnesium sulfate on vasogenic brain edema formation and blood-brain barrier (BBB) disruption caused by ischemia-reperfusion (IR) in a rat model of ischemic stroke. A total of 72 male Sprague-Dawley rats were categorized into the following three primary groups: sham, control ischemic, magnesium-sulfate-treated (300 mg/kg loading dose, followed by an additional 100 mg/kg) ischemic (n = 24 in each group). Transient focal cerebral ischemia was induced by 60-min-long occlusion of the left middle cerebral artery, followed by 24-h-long reperfusion. Sensorimotor deficits, infarct volume, and brain edema were evaluated at the end of the reperfusion period. The BBB permeability was assessed by Evans Blue extravasation technique. Lipid peroxidation levels were assessed by measuring the malondialdehyde content in the brain tissue homogenate, and the activities of the antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase were detected according to the technical manual of the assay kits. Induction of cerebral ischemia in the control group produced considerable BBB damage in conjunction with severe brain edema formation. Treatment with magnesium sulfate significantly attenuated brain edema and protected BBB integrity in the ischemic lesioned hemisphere. In addition, magnesium sulfate reduced lipid peroxidation and increased antioxidant protection of brain tissue by upregulating the activities of antioxidant enzymes. Treatment with magnesium sulfate protected BBB integrity against IR-induced damage and reduced vasogenic edema formation partly via antioxidant mechanisms in a rat model of acute ischemic stroke.
Collapse
Affiliation(s)
- Javad Shadman
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nooshin Sadeghian
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Moradi
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shahab Bohlooli
- Department of Pharmacology, Pharmacy School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamdollah Panahpour
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran.
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
20
|
Cerebral Edema and Intracranial Pressure in the Neurocritical Care Unit. Neurocrit Care 2019. [DOI: 10.1017/9781107587908.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
21
|
Calcitriol protects the Blood-Brain Barrier integrity against ischemic stroke and reduces vasogenic brain edema via antioxidant and antiapoptotic actions in rats. Brain Res Bull 2019; 150:281-289. [PMID: 31220552 DOI: 10.1016/j.brainresbull.2019.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND Vasogenic brain edema is the most important complication of ischemic stroke that aggravates primary brain injury. Ischemia-Reperfusion (IR)-induced Blood-Brain Barrier (BBB) impairment limits the use of recombinant tissue plasminogen activator (r-tPA) by increasing the possibility of hemorrhagic transformation and contributing to vasogenic edema and neuroinflammation. This study examined the effects of post-ischemic treatment with calcitriol on cerebral infarction, vasogenic edema formation and BBB disruption in a rat model of ischemic stroke. METHODS Male Sprague-Dawley rats were divided into three main groups, including the sham, IR + vehicle and IR + calcitriol groups. Transient focal cerebral ischemia was induced by a 60-min-long occlusion of the left middle cerebral artery. The infarct volume, brain edema, BBB permeability and antioxidant enzyme activities were evaluated 24 h after ischemia. Immunohistochemical analysis was conducted to investigate cell apoptosis and Brain-Derived Neurotrophic Factor (BDNF) protein expression five days after ischemia. RESULTS Compared to the IR + vehicle group, the IR + calcitriol group showed a reduced brain infarction volume, attenuated brain edema formation and improved BBB function. These protective effects were followed by the upregulation of antioxidant enzyme activities in the brain tissue. Additionally, a diminished cell apoptosis and an increased BDNF immunoreactivity were obtained in the IR + calcitriol group. CONCLUSION Calcitriol may reduce brain injury and attenuate vasogenic edema by upregulating antioxidant enzymes activities, reducing cell apoptosis and increasing BDNF protein in the brain tissue in a rat model of ischemic stroke.
Collapse
|
22
|
Luo C, Bian X, Zhang Q, Xia Z, Liu B, Chen Q, Ke C, Wu JL, Zhao Y. Shengui Sansheng San Ameliorates Cerebral Energy Deficiency via Citrate Cycle After Ischemic Stroke. Front Pharmacol 2019; 10:386. [PMID: 31065240 PMCID: PMC6489525 DOI: 10.3389/fphar.2019.00386] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/28/2019] [Indexed: 12/13/2022] Open
Abstract
Cerebral energy deficiency is a key pathophysiologic cascade that results in neuronal injury and necrosis after ischemic stroke. Shengui Sansheng San (SSS) has been used to treat stroke for more than 300 years. In present study, we investigated the therapeutic efficacy and mechanism of SSS extraction on cerebral energy deficiency post-stroke. In permanent middle cerebral artery occlusion (pMCAo) model of rats, it suggested that SSS extraction in dose-dependent manner improved neurological function, cerebral blood flow (CBF), 18F-2-deoxy-glucose uptake and the density and diameter of alpha smooth muscle actin (α-SMA) positive vasculature in ipsilateral area, as well as decreased infarcted volume. Meanwhile, the metabolomics study in cerebrospinal fluid (CSF) was performed by using 5-(diisopropylamino)amylamine (DIAAA) derivatization-UHPLC-Q-TOF/MS approach. Eighty-eight endogenous metabolites were identified, and mainly involved in citrate cycle, fatty acid biosynthesis, aminoacyl-tRNA biosynthesis, amino acids metabolism and biosynthesis, etc. The remarkable increase of citrate in CSF after treatment with three dosages indicated that the therapeutic mechanism of SSS extraction might be related with citrate cycle. Simultaneously, it showed that high dosage group significantly increased peripheral blood glucose level, the expressions of glucose transporter (GLUT) 1, GLUT3, and monocarboxylic acid transporter 1 (MCT1), which contributed to the transportation of glucose and lactate. By the regulations of phosphorylated pyruvate dehydrogenase E1-alpha (p-PDHA1), acetyl CoA synthetase and citrate synthetase (CS), the levels of citrate and its upstream molecules (pyruvate and acetyl CoA) in peri-infarction zone further enhanced, which ultimately caused the massive yield of adenosine triphosphate (ATP). Our study first demonstrated that SSS extraction could ameliorate cerebral energy deficiency after ischemia by citrate cycle, which is characterized by the enhancements of glucose supply, transportation, utilization, and metabolism.
Collapse
Affiliation(s)
- Cheng Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xiqing Bian
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Qian Zhang
- Department of Biotherapy, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Zhenyan Xia
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Bowen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Qi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chienchih Ke
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.,Biomedical Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Jian-Lin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
23
|
Zhou Z, Ikegaya Y, Koyama R. The Astrocytic cAMP Pathway in Health and Disease. Int J Mol Sci 2019; 20:E779. [PMID: 30759771 PMCID: PMC6386894 DOI: 10.3390/ijms20030779] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are major glial cells that play critical roles in brain homeostasis. Abnormalities in astrocytic functions can lead to brain disorders. Astrocytes also respond to injury and disease through gliosis and immune activation, which can be both protective and detrimental. Thus, it is essential to elucidate the function of astrocytes in order to understand the physiology of the brain to develop therapeutic strategies against brain diseases. Cyclic adenosine monophosphate (cAMP) is a major second messenger that triggers various downstream cellular machinery in a wide variety of cells. The functions of astrocytes have also been suggested as being regulated by cAMP. Here, we summarize the possible roles of cAMP signaling in regulating the functions of astrocytes. Specifically, we introduce the ways in which cAMP pathways are involved in astrocyte functions, including (1) energy supply, (2) maintenance of the extracellular environment, (3) immune response, and (4) a potential role as a provider of trophic factors, and we discuss how these cAMP-regulated processes can affect brain functions in health and disease.
Collapse
Affiliation(s)
- Zhiwen Zhou
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan.
- Center for Information and Neural Networks, Suita City, Osaka 565-0871, Japan.
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
24
|
Hada B, Karmacharya MB, Park SR, Choi BH. Low-Intensity Ultrasound Decreases Ischemia-Induced Edema by Inhibiting N-Methyl-d-Aspartic Acid Receptors. Can J Neurol Sci 2018; 45:675-681. [PMID: 30430968 DOI: 10.1017/cjn.2018.331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND We have previously shown that low-intensity ultrasound (LIUS), a noninvasive mechanical stimulus, inhibits brain edema formation induced by oxygen and glucose deprivation (OGD) or treatment with glutamate, a mediator of OGD-induced edema, in acute rat hippocampal slice model in vitro. METHODS In this study, we treated the rat hippocampal slices with N-methyl-d-aspartic acid (NMDA) or (S)-3,5-dihydroxyphenylglycine (DHPG) to determine whether these different glutamate receptor agonists induce edema. The hippocampal slices were then either sonicated with LIUS or treated with N-methyl-d-aspartic acid receptor (NMDAR) antagonists, namely, MK-801 and ketamine, and observed their effects on edema formation. RESULTS We observed that treatment with NMDA, an agonist of ionotropic glutamate receptors, induced brain edema at similar degrees compared with that induced by OGD. However, treatment with DHPG, an agonist of metabotropic glutamate receptors, did not significantly induce brain edema. Treatment with the NMDAR antagonists MK-801 or ketamine efficiently prevented brain edema formation by both OGD and NMDA in a concentration-dependent manner. N-Methyl-d-aspartic acid-induced brain edema was alleviated by LIUS in an intensity-dependent manner when ultrasound was administered at 30, 50, or 100 mW/cm2 for 20 minutes before the induction of the edema. Furthermore, LIUS reduced OGD- and NMDA-induced phosphorylation of NMDARs at Y1325. CONCLUSION These results suggest that LIUS can inhibit OGD- or NMDA-induced NMDAR activation by preventing NMDAR phosphorylation, thereby reducing a subsequent brain edema formation. The mechanisms by which LIUS inhibits NMDAR phosphorylation need further investigation.
Collapse
Affiliation(s)
- Binika Hada
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | | | - So R Park
- Department of Physiology and Biophysics, Inha University College of Medicine, Incheon, Korea
| | - Byung H Choi
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
25
|
Bylicky MA, Mueller GP, Day RM. Mechanisms of Endogenous Neuroprotective Effects of Astrocytes in Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6501031. [PMID: 29805731 PMCID: PMC5901819 DOI: 10.1155/2018/6501031] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/19/2018] [Indexed: 12/11/2022]
Abstract
Astrocytes, once believed to serve only as "glue" for the structural support of neurons, have been demonstrated to serve critical functions for the maintenance and protection of neurons, especially under conditions of acute or chronic injury. There are at least seven distinct mechanisms by which astrocytes protect neurons from damage; these are (1) protection against glutamate toxicity, (2) protection against redox stress, (3) mediation of mitochondrial repair mechanisms, (4) protection against glucose-induced metabolic stress, (5) protection against iron toxicity, (6) modulation of the immune response in the brain, and (7) maintenance of tissue homeostasis in the presence of DNA damage. Astrocytes support these critical functions through specialized responses to stress or toxic conditions. The detoxifying activities of astrocytes are essential for maintenance of the microenvironment surrounding neurons and in whole tissue homeostasis. Improved understanding of the mechanisms by which astrocytes protect the brain could lead to the development of novel targets for the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Michelle A. Bylicky
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Gregory P. Mueller
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
26
|
Abstract
Oncotic cell death or oncosis represents a major mechanism of cell death in ischaemic stroke, occurring in many central nervous system (CNS) cell types including neurons, glia and vascular endothelial cells. In stroke, energy depletion causes ionic pump failure and disrupts ionic homeostasis. Imbalance between the influx of Na+ and Cl- ions and the efflux of K+ ions through various channel proteins and transporters creates a transmembrane osmotic gradient, with ensuing movement of water into the cells, resulting in cell swelling and oncosis. Oncosis is a key mediator of cerebral oedema in ischaemic stroke, contributing directly through cytotoxic oedema, and indirectly through vasogenic oedema by causing vascular endothelial cell death and disruption of the blood-brain barrier (BBB). Hence, inhibition of uncontrolled ionic flux represents a novel and powerful strategy in achieving neuroprotection in stroke. In this review, we provide an overview of oncotic cell death in the pathology of stroke. Importantly, we summarised the therapeutically significant pathways of water, Na+, Cl- and K+ movement across cell membranes in the CNS and their respective roles in the pathobiology of stroke.
Collapse
|
27
|
Woo CW, Kwon JI, Kim KW, Kim JK, Jeon SB, Jung SC, Choi CG, Kim ST, Kim J, Ham SJ, Shim WH, Sung YS, Ha HK, Choi Y, Woo DC. The administration of hydrogen sulphide prior to ischemic reperfusion has neuroprotective effects in an acute stroke model. PLoS One 2017; 12:e0187910. [PMID: 29161281 PMCID: PMC5697867 DOI: 10.1371/journal.pone.0187910] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 10/28/2017] [Indexed: 11/18/2022] Open
Abstract
Emerging evidence has suggested that hydrogen sulfide (H2S) may alleviate the cellular damage associated with cerebral ischemia/reperfusion (I/R) injury. In this study, we assessed using 1H-magnetic resonance imaging/magnetic resonance spectroscopy (1H-MRI/MRS) and histologic analysis whether H2S administration prior to reperfusion has neuroprotective effects. We also evaluated for differences in the effects of H2S treatment at 2 time points. 1H-MRI/MRS data were obtained at baseline, and at 3, 9, and 24 h after ischemia from 4 groups: sham, control (I/R injury), sodium hydrosulfide (NaHS)-30 and NaHS-1 (NaHS delivery at 30 and 1 min before reperfusion, respectively). The total infarct volume and the midline shift at 24 h post-ischemia were lowest in the NaHS-1, followed by the NaHS-30 and control groups. Peri-infarct volume was significantly lower in the NaHS-1 compared to NaHS-30 and control animals. The relative apparent diffusion coefficient (ADC) in the peri-infarct region showed that the NaHS-1 group had significantly lower values compared to the NaHS-30 and control animals and that NaHS-1 rats showed significantly higher relative T2 values in the peri-infarct region compared to the controls. The relative ADC value, relative T2 value, levels of N-acetyl-L-aspartate (NAA), and the NAA, glutamate, and taurine combination score (NGT) in the ischemic core region at 24 h post-ischemia did not differ significantly between the 2 NaHS groups and the control except that the NAA and NGT values were higher in the peri-infarct region of the NaHS-1 animals at 9 h post-ischemia. In the ischemic core and peri-infarct regions, the apoptosis rate was lowest in the NaHS-1 group, followed by the NaHS-30 and control groups. Our results suggest that H2S treatment has neuroprotective effects on the peri-infarct region during the evolution of I/R injury. Furthermore, our findings indicate that the administration of H2S immediately prior to reperfusion produces the highest neuroprotective effects.
Collapse
Affiliation(s)
- Chul-Woong Woo
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Jae-Im Kwon
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Kyung-Won Kim
- Department of Radiology, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Jeong-Kon Kim
- Department of Radiology, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Sang-Beom Jeon
- Department of Neurology, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Seung-Chae Jung
- Department of Radiology, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Choong-Gon Choi
- Department of Radiology, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Sang-Tae Kim
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Jinil Kim
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Su Jeong Ham
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Woo-Hyun Shim
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Yu Sub Sung
- Department of Radiology, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Hyun Kwon Ha
- Medical Research Institute, Gangneung Asan Hospital Gangneung-si, Gangwon-do, Republic of Korea
| | - Yoonseok Choi
- Medical Research Institute, Gangneung Asan Hospital Gangneung-si, Gangwon-do, Republic of Korea
- * E-mail: (YSC); (DCW)
| | - Dong-Cheol Woo
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
- * E-mail: (YSC); (DCW)
| |
Collapse
|
28
|
Ardelt AA, Carpenter RS, Iwuchukwu I, Zhang A, Lin W, Kosciuczuk E, Hinkson C, Rebeiz T, Reitz S, King PH. Transgenic expression of HuR increases vasogenic edema and impedes functional recovery in rodent ischemic stroke. Neurosci Lett 2017; 661:126-131. [PMID: 28982595 DOI: 10.1016/j.neulet.2017.09.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND PURPOSE Ischemic stroke produces significant morbidity and mortality, and acute interventions are limited by short therapeutic windows. Novel approaches to neuroprotection and neurorepair are necessary. HuR is an RNA-binding protein (RBP) which modulates RNA stability and translational efficiency of genes linked to ischemic stroke injury. METHODS Using a transgenic (Tg) mouse model, we examined the impact of ectopic HuR expression in astrocytes on acute injury evolution after transient middle cerebral artery occlusion (tMCAO). RESULTS HuR transgene expression was detected in astrocytes in perilesional regions and contralaterally. HuR Tg mice did not improve neurologically 72h after injury, whereas littermate controls did. In Tg mice, increased cerebral vascular permeability and edema were observed. Infarct volume was not affected by the presence of the transgene. CONCLUSIONS Ectopic expression of HuR in astrocytes worsens outcome after transient ischemic stroke in mice in part by increasing vasogenic cerebral edema. These findings suggest that HuR could be a therapeutic target in cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Agnieszka A Ardelt
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - Randall S Carpenter
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - Ifeanyi Iwuchukwu
- Department of Neurocritical Care, Ochsner Medical Center, 1514 Jefferson Hwy., New Orleans, LA 70121, United States.
| | - An Zhang
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - William Lin
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - Ewa Kosciuczuk
- Division of Hematology-Oncology, Northwestern University, 675 North St. Clair, Chicago, IL 60611, United States.
| | - Cyrus Hinkson
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - Tania Rebeiz
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - Sydney Reitz
- Department of Neurology, University of Chicago, 5841 S. Maryland Ave, MC2030, Chicago, IL 60637, United States.
| | - Peter H King
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, United States.
| |
Collapse
|
29
|
Ilesanmi OB, Akinmoladun AC, Olayeriju OS, Saliu IO, Olaleye MT, Akindahunsi AA. MODULATION OF KEY BIOCHEMICAL MARKERS RELEVANT TO STROKE BY ANTIARIS AFRICANA LEAF EXTRACT FOLLOWING CEREBRAL ISCHEMIA/REPERFUSION INJURY. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2017; 14:253-264. [PMID: 28638888 PMCID: PMC5471473 DOI: 10.21010/ajtcam.v14i4.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND Oxidative stress plays a significant role in stroke pathogenesis. Hence, plants rich in antioxidant phytochemicals have been suggested as effective remedies for prevention and treatment of stroke and other neurological diseases. Antiaris africana Engl. (Moraceae) is traditionally used for the management of brain-related problems but there is paucity of data on its anti-stroke potential. MATERIALS AND METHODS Ischemia/reperfusion injury was induced by a 30 min bilateral common carotid artery occlusion/ 2 h reperfusion (BCCAO/R) in the brain of male Wistar rats. A sham-operated group which was not subjected to BCCAO/R and a group subjected to BCCAO/R without treatment with MEA served as controls. The ameliorative effect of 14 days of pretreatment with 50 mg/kg or 100 mg/kg A. africana methanol leaf extract (MEA) on BCCAO/R-mediated alterations to key markers of oxidative stress (malondialdehyde, reduced glutathione, xanthine oxidase, superoxide dismutase, catalase and glutathione peroxidase) and neurochemical disturbances and excitotoxicity (myeloperoxidase, glutamine synthetase, Na+/K+ ATPase, acetylcholinesterase and tyrosine hydroxylase), was evaluated and compared with the effect produced by treatment with 20 mg/kg quercetin as a reference standard. RESULTS Results show that pretreatment with MEA significantly mitigated or reversed BCCAO/R-induced changes in the level or activity of the evaluated biochemical markers of oxidative stress, neurochemical dysfunction and excitotoxicity compared with the BCCAO/R untreated control group (p < 0.05). The effect produced by 100 mg/kg MEA was similar to that of the reference standard, quercetin. CONCLUSION These results revealed the neuroprotective potential of A. africana in stroke and other ischemia-related pathologies.
Collapse
Affiliation(s)
- Omotayo B. Ilesanmi
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology, PMB 704, Akure, Nigeria
- Department of Biological Sciences, Faculty of Science, Federal University Otuoke, Otuoke, Nigeria
| | - Afolabi C. Akinmoladun
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology, PMB 704, Akure, Nigeria
| | - Olanrewaju Sam Olayeriju
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology, PMB 704, Akure, Nigeria
| | - Ibrahim Olabayode Saliu
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology, PMB 704, Akure, Nigeria
| | - M. Tolulope Olaleye
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology, PMB 704, Akure, Nigeria
| | - Afolabi A. Akindahunsi
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology, PMB 704, Akure, Nigeria
| |
Collapse
|
30
|
Sifat AE, Vaidya B, Abbruscato TJ. Blood-Brain Barrier Protection as a Therapeutic Strategy for Acute Ischemic Stroke. AAPS JOURNAL 2017; 19:957-972. [PMID: 28484963 DOI: 10.1208/s12248-017-0091-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/18/2017] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) is a vital component of the neurovascular unit (NVU) containing tight junctional (TJ) proteins and different ion and nutrient transporters which maintain normal brain physiology. BBB disruption is a major pathological hallmark in the course of ischemic stroke which is regulated by the actions of different factors working at different stages of cerebral ischemia including matrix metalloproteinases (MMPs), inflammatory modulators, vesicular trafficking, oxidative pathways, and junctional-cytoskeletal interactions. These components interact further to disrupt maintenance of both the paracellular and transport barriers of the central nervous system (CNS) to worsen ischemic brain injury and the propensity for hemorrhagic transformation (HT) associated with injury and/or thrombolytic therapy with tissue-type plasminogen activator (tPA). We propose that these complex molecular pathways should be evaluated further so that they could be targeted alone or in combination to protect the BBB during cerebral ischemia. These types of novel interventions should be guided by advanced imaging techniques for better diagnosis of BBB damage which may exert significant therapeutic benefit including the extension of therapeutic window of tPA. This review will focus on the different stages and mechanisms of BBB damage in acute ischemic stroke and novel therapeutic strategies to target those pathways for better therapeutic outcome in stroke.
Collapse
Affiliation(s)
- Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter, Amarillo, Texas, 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter, Amarillo, Texas, 79106, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter, Amarillo, Texas, 79106, USA.
| |
Collapse
|
31
|
Babadjouni RM, Walcott BP, Liu Q, Tenser MS, Amar AP, Mack WJ. Neuroprotective delivery platforms as an adjunct to mechanical thrombectomy. Neurosurg Focus 2017; 42:E4. [PMID: 28366053 DOI: 10.3171/2017.1.focus16514] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite the success of numerous neuroprotective strategies in animal and preclinical stroke models, none have effectively translated to clinical medicine. A multitude of influences are likely responsible. Two such factors are inefficient recanalization strategies for large vessel occlusions and suboptimal delivery methods/platforms for neuroprotective agents. The recent endovascular stroke trials have established a new paradigm for large vessel stroke treatment. The associated advent of advanced mechanical revascularization devices and new stroke technologies help address each of these existing gaps. A strategy combining effective endovascular revascularization with administration of neuroprotective therapies is now practical and could have additive, if not synergistic, effects. This review outlines past and current neuroprotective strategies assessed in acute stroke trials. The discussion focuses on delivery platforms and their potential applicability to endovascular stoke treatment.
Collapse
Affiliation(s)
| | - Brian P Walcott
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Matthew S Tenser
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Arun P Amar
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - William J Mack
- Zilkha Neurogenetic Institute and.,Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
32
|
Phase I and Phase II Therapies for Acute Ischemic Stroke: An Update on Currently Studied Drugs in Clinical Research. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4863079. [PMID: 28286764 PMCID: PMC5329656 DOI: 10.1155/2017/4863079] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/23/2016] [Indexed: 02/06/2023]
Abstract
Acute ischemic stroke is a devastating cause of death and disability, consequences of which depend on the time from ischemia onset to treatment, the affected brain region, and its size. The main targets of ischemic stroke therapy aim to restore tissue perfusion in the ischemic penumbra in order to decrease the total infarct area by maintaining blood flow. Advances in research of pathological process and pathways during acute ischemia have resulted in improvement of new treatment strategies apart from restoring perfusion. Additionally, limiting the injury severity by manipulating the molecular mechanisms during ischemia has become a promising approach, especially in animal research. The purpose of this article is to review completed and ongoing phases I and II trials for the treatment of acute ischemic stroke, reviewing studies on antithrombotic, thrombolytic, neuroprotective, and antineuroinflammatory drugs that may translate into more effective treatments.
Collapse
|
33
|
Al-Attar R, Zhang Y, Storey KB. Osmolyte regulation by TonEBP/NFAT5 during anoxia-recovery and dehydration-rehydration stresses in the freeze-tolerant wood frog ( Rana sylvatica). PeerJ 2017; 5:e2797. [PMID: 28133564 PMCID: PMC5251939 DOI: 10.7717/peerj.2797] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 11/15/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The wood frog, Rana sylvatica, tolerates freezing as a means of winter survival. Freezing is considered to be an ischemic/anoxic event in which oxygen delivery is significantly impaired. In addition, cellular dehydration occurs during freezing because water is lost to extracellular compartments in order to promote freezing. In order to prevent severe cell shrinkage and cell death, it is important for the wood frog to have adaptive mechanisms for osmoregulation. One important mechanism of cellular osmoregulation occurs through the cellular uptake/production of organic osmolytes like sorbitol, betaine, and myo-inositol. Betaine and myo-inositol are transported by the proteins BGT-1 and SMIT, respectively. Sorbitol on the other hand, is synthesized inside the cell by the enzyme aldose reductase. These three proteins are regulated at the transcriptional level by the transcription factor, NFAT5/TonEBP. Therefore, the objective of this study was to elucidate the role of NFAT5/TonEBP in regulating BGT-1, SMIT, and aldose reductase, during dehydration and anoxia in the wood frog muscle, liver, and kidney tissues. METHODS Wood frogs were subjected to 24 h anoxia-4 h recovery and 40% dehydration-full rehydration experiments. Protein levels of NFAT5, BGT-1, SMIT, and aldose reductase were studied using immunoblotting in muscle, liver, and kidney tissues. RESULTS Immunoblotting results demonstrated downregulations in NFAT5 protein levels in both liver and kidney tissues during anoxia (decreases by 41% and 44% relative to control for liver and kidney, respectively). Aldose reductase protein levels also decreased in both muscle and kidney tissues during anoxia (by 37% and 30% for muscle and kidney, respectively). On the other hand, BGT-1 levels increased during anoxia in muscle (0.9-fold compared to control) and kidney (1.1-fold). Under 40% dehydration, NFAT5 levels decreased in liver by 53%. Aldose reductase levels also decreased by 42% in dehydrated muscle, and by 35% in dehydrated liver. In contrast, BGT-1 levels increased by 1.4-fold in dehydrated liver. SMIT levels also increased in both dehydrated muscle and liver (both by 0.8-fold). DISCUSSION Overall, we observed that osmoregulation through an NFAT5-mediated pathway is both tissue- and stress-specific. In both anoxia and dehydration, there appears to be a general reduction in NFAT5 levels resulting in decreased aldose reductase levels, however BGT-1 and SMIT levels still increase in certain tissues. Therefore, the regulation of osmoregulatory genes during dehydration and anoxia occurs beyond the transcriptional level, and it possibly involves RNA processing as well. These novel findings on the osmoregulatory mechanisms utilized by the wood frog advances our knowledge of osmoregulation during anoxia and dehydration. In addition, these findings highlight the importance of using this model to study molecular adaptations during stress.
Collapse
Affiliation(s)
- Rasha Al-Attar
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University , Ottawa , ON , Canada
| | - Yichi Zhang
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University , Ottawa , ON , Canada
| | - Kenneth B Storey
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University , Ottawa , ON , Canada
| |
Collapse
|
34
|
|
35
|
Targeting antioxidant enzyme expression as a therapeutic strategy for ischemic stroke. Neurochem Int 2016; 107:23-32. [PMID: 28043837 DOI: 10.1016/j.neuint.2016.12.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/05/2016] [Accepted: 12/17/2016] [Indexed: 11/24/2022]
Abstract
During ischemic stroke, neurons and glia are subjected to damage during the acute and neuroinflammatory phases of injury. Production of reactive oxygen species (ROS) from calcium dysregulation in neural cells and the invasion of activated immune cells are responsible for stroke-induced neurodegeneration. Scientists have failed thus far to identify antioxidant-based drugs that can enhance neural cell survival and improve recovery after stroke. However, several groups have demonstrated success in protecting against stroke by increasing expression of antioxidant enzymes in neural cells. These enzymes, which include but are not limited to enzymes in the glutathione peroxidase, catalase, and superoxide dismutase families, degrade ROS that otherwise damage cellular components such as DNA, proteins, and lipids. Several groups have identified cellular therapies including neural stem cells and human umbilical cord blood cells, which exert neuroprotective and oligoprotective effects through the release of pro-survival factors that activate PI3K/Akt signaling to upregulation of antioxidant enzymes. Other studies demonstrate that treatment with soluble factors released by these cells yield similar changes in enzyme expression after stroke. Treatment with the cytokine leukemia inhibitory factor increases the expression of peroxiredoxin IV and metallothionein III in glia and boosts expression of superoxide dismutase 3 in neurons. Through cell-specific upregulation of these enzymes, LIF and other Akt-inducing factors have the potential to protect multiple cell types against damage from ROS during the early and late phases of ischemic damage.
Collapse
|
36
|
Wang H, Li P, Xu N, Zhu L, Cai M, Yu W, Gao Y. Paradigms and mechanisms of inhalational anesthetics mediated neuroprotection against cerebral ischemic stroke. Med Gas Res 2016; 6:194-205. [PMID: 28217291 PMCID: PMC5223310 DOI: 10.4103/2045-9912.196901] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cerebral ischemic stroke is a leading cause of serious long-term disability and cognitive dysfunction. The high mortality and disability of cerebral ischemic stroke is urging the health providers, including anesthesiologists and other perioperative professioners, to seek effective protective strategies, which are extremely limited, especially for those perioperative patients. Intriguingly, several commonly used inhalational anesthetics are recently suggested to possess neuroprotective effects against cerebral ischemia. This review introduces multiple paradigms of inhalational anesthetic treatments that have been investigated in the setting of cerebral ischemia, such as preconditioning, proconditioning and postconditioning with a variety of inhalational anesthetics. The pleiotropic mechanisms underlying these inhalational anesthetics-afforded neuroprotection against stroke are also discussed in detail, including the common pathways shared by most of the inhalational anesthetic paradigms, such as anti-excitotoxicity, anti-apoptosis and anti-inflammation. There are also distinct mechanisms involved in specific paradigms, such as preserving blood brain barrier integrity, regulating cerebral blood flow and catecholamine release. The ready availability of these inhalational anesthetics bedside and renders them a potentially translatable stroke therapy attracting great efforts for understanding of the underlying mechanisms.
Collapse
Affiliation(s)
- Hailian Wang
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peiying Li
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Na Xu
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ling Zhu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mengfei Cai
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanqin Gao
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Hertz L, Chen Y. Importance of astrocytes for potassium ion (K+) homeostasis in brain and glial effects of K+ and its transporters on learning. Neurosci Biobehav Rev 2016; 71:484-505. [DOI: 10.1016/j.neubiorev.2016.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/12/2016] [Accepted: 09/23/2016] [Indexed: 10/20/2022]
|
38
|
Multifactorial Effects on Different Types of Brain Cells Contribute to Ammonia Toxicity. Neurochem Res 2016; 42:721-736. [PMID: 27286679 DOI: 10.1007/s11064-016-1966-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022]
Abstract
Effects of ammonia on astrocytes play a major role in hepatic encephalopathy, acute liver failure and other diseases caused by increased arterial ammonia concentrations (e.g., inborn errors of metabolism, drug or mushroom poisoning). There is a direct correlation between arterial ammonia concentration, brain ammonia level and disease severity. However, the pathophysiology of hyperammonemic diseases is disputed. One long recognized factor is that increased brain ammonia triggers its own detoxification by glutamine formation from glutamate. This is an astrocytic process due to the selective expression of the glutamine synthetase in astrocytes. A possible deleterious effect of the resulting increase in glutamine concentration has repeatedly been discussed and is supported by improvement of some pathologic effects by GS inhibition. However, this procedure also inhibits a large part of astrocytic energy metabolism and may prevent astrocytes from responding to pathogenic factors. A decrease of the already low glutamate concentration in astrocytes due to increased synthesis of glutamine inhibits the malate-aspartate shuttle and energy metabolism. A more recently described pathogenic factor is the resemblance between NH4+ and K+ in their effects on the Na+,K+-ATPase and the Na+,K+, 2 Cl- and water transporter NKCC1. Stimulation of the Na+,K+-ATPase driven NKCC1 in both astrocytes and endothelial cells is essential for the development of brain edema. Na+,K+-ATPase stimulation also activates production of endogenous ouabains. This leads to oxidative and nitrosative damage and sensitizes NKCC1. Administration of ouabain antagonists may accordingly have therapeutic potential in hyperammonemic diseases.
Collapse
|
39
|
Abstract
Stroke is the second foremost cause of mortality worldwide and a major cause of long-term disability. Due to changes in lifestyle and an aging population, the incidence of stroke continues to increase and stroke mortality predicted to exceed 12 % by the year 2030. However, the development of pharmacological treatments for stroke has failed to progress much in over 20 years since the introduction of the thrombolytic drug, recombinant tissue plasminogen activator. These alarming circumstances caused many research groups to search for alternative treatments in the form of neuroprotectants. Here, we consider the potential use of phytochemicals in the treatment of stroke. Their historical use in traditional medicine and their excellent safety profile make phytochemicals attractive for the development of therapeutics in human diseases. Emerging findings suggest that some phytochemicals have the ability to target multiple pathophysiological processes involved in stroke including oxidative stress, inflammation and apoptotic cell death. Furthermore, epidemiological studies suggest that the consumption of plant sources rich in phytochemicals may reduce stroke risk, and so reinforce the possibility of developing preventative or neuroprotectant therapies for stroke. In this review, we describe results of preclinical studies that demonstrate beneficial effects of phytochemicals in experimental models relevant to stroke pathogenesis, and we consider their possible mechanisms of action.
Collapse
|
40
|
Wang YF, Parpura V. Central Role of Maladapted Astrocytic Plasticity in Ischemic Brain Edema Formation. Front Cell Neurosci 2016; 10:129. [PMID: 27242440 PMCID: PMC4865516 DOI: 10.3389/fncel.2016.00129] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/29/2016] [Indexed: 12/24/2022] Open
Abstract
Brain edema formation and the ensuing brain damages are the major cause of high mortality and long term disability following the occurrence of ischemic stroke. In this process, oxygen and glucose deprivation and the resulting reperfusion injury play primary roles. In response to the ischemic insult, the neurovascular unit experiences both intracellular and extracellular edemas, associated with maladapted astrocytic plasticity. The astrocytic plasticity includes both morphological and functional plasticity. The former involves a reactive gliosis and the subsequent glial retraction. It relates to the capacity of astrocytes to buffer changes in extracellular chemical levels, particularly K+ and glutamate, as well as the integrity of the blood-brain barrier (BBB). The latter involves the expression and activity of a series of ion and water transport proteins. These molecules are grouped together around glial fibrillary acidic protein (GFAP) and water channel protein aquaporin 4 (AQP4) to form functional networks, regulate hydromineral balance across cell membranes and maintain the integrity of the BBB. Intense ischemic challenges can disrupt these capacities of astrocytes and result in their maladaptation. The maladapted astrocytic plasticity in ischemic stroke cannot only disrupt the hydromineral homeostasis across astrocyte membrane and the BBB, but also leads to disorders of the whole neurovascular unit. This review focuses on how the maladapted astrocytic plasticity in ischemic stroke plays the central role in the brain edema formation.
Collapse
Affiliation(s)
- Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University Harbin, China
| | - Vladimir Parpura
- Department of Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
41
|
Liu Y, Wang W, Li Y, Xiao Y, Cheng J, Jia J. The 5-Lipoxygenase Inhibitor Zileuton Confers Neuroprotection against Glutamate Oxidative Damage by Inhibiting Ferroptosis. Biol Pharm Bull 2016; 38:1234-9. [PMID: 26235588 DOI: 10.1248/bpb.b15-00048] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
5-Lipoxygenase (5-LOX) inhibitors have been shown to be protective in several neurodegenerative disease models; however, the underlying mechanisms remain unclear. We investigated whether 5-LOX inhibitor zileuton conferred direct neuroprotection against glutamate oxidative toxicity by inhibiting ferroptosis, a newly identified iron-dependent programmed cell death. Treatment of HT22 mouse neuronal cell line with glutamate resulted in significant cell death, which was inhibited by zileuton in a dose-dependent manner. Consistently, zileuton decreased glutamate-induced production of reactive oxygen species but did not restore glutamate-induced depletion of glutathione. Moreover, the pan-caspase inhibitor Z-Val-Ala-Asp(OMe)-fluoromethyl ketone (ZVAD-fmk) neither prevented HT22 cell death induced by glutamate nor affected zileuton protection against glutamate oxidative toxicity, suggesting that zileuton did not confer neuroprotection by inhibiting caspase-dependent apoptosis. Interestingly, glutamate-induced HT22 cell death was significantly inhibited by the ferroptosis inhibitor ferrostatin-1. Moreover, zileuton protected HT22 neuronal cells from erastin-induced ferroptosis. However, we did not observe synergic protective effects of zileuton and ferrostatin-1 on glutamate-induced cell death. These results suggested that both the 5-LOX inhibitor zileuton and the ferropotosis inhibitor ferrostatin-1 acted through the same cascade to protect against glutamate oxidative toxicity. In conclusion, our results suggested that zileuton protected neurons from glutamate-induced oxidative stress at least in part by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Yang Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Soochow University
| | | | | | | | | | | |
Collapse
|
42
|
Kuzmin NV, Wesseling P, Hamer PCDW, Noske DP, Galgano GD, Mansvelder HD, Baayen JC, Groot ML. Third harmonic generation imaging for fast, label-free pathology of human brain tumors. BIOMEDICAL OPTICS EXPRESS 2016; 7:1889-904. [PMID: 27231629 PMCID: PMC4871089 DOI: 10.1364/boe.7.001889] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/08/2016] [Accepted: 03/12/2016] [Indexed: 05/07/2023]
Abstract
In brain tumor surgery, recognition of tumor boundaries is key. However, intraoperative assessment of tumor boundaries by the neurosurgeon is difficult. Therefore, there is an urgent need for tools that provide the neurosurgeon with pathological information during the operation. We show that third harmonic generation (THG) microscopy provides label-free, real-time images of histopathological quality; increased cellularity, nuclear pleomorphism, and rarefaction of neuropil in fresh, unstained human brain tissue could be clearly recognized. We further demonstrate THG images taken with a GRIN objective, as a step toward in situ THG microendoscopy of tumor boundaries. THG imaging is thus a promising tool for optical biopsies.
Collapse
Affiliation(s)
- N. V. Kuzmin
- LaserLab Amsterdam, VU University, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - P. Wesseling
- Dept. of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Dept. of Pathology, Radboud University Medical Center, Geert Grooteplein Zuid, 6525 GA Nijmegen, The Netherlands
- Amsterdam Brain Tumor Center, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - P. C. de Witt Hamer
- Dept. of Neurosurgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Brain Tumor Center, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - D. P. Noske
- Dept. of Neurosurgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Brain Tumor Center, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - G. D. Galgano
- LaserLab Amsterdam, VU University, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
| | - H. D. Mansvelder
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - J. C. Baayen
- Dept. of Neurosurgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - M. L. Groot
- LaserLab Amsterdam, VU University, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
43
|
Jia SW, Liu XY, Wang SC, Wang YF. Vasopressin Hypersecretion-Associated Brain Edema Formation in Ischemic Stroke: Underlying Mechanisms. J Stroke Cerebrovasc Dis 2016; 25:1289-300. [PMID: 27068863 DOI: 10.1016/j.jstrokecerebrovasdis.2016.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Brain edema formation is a major cause of brain damages and the high mortality of ischemic stroke. The aim of this review is to explore the relationship between ischemic brain edema formation and vasopressin (VP) hypersecretion in addition to the oxygen and glucose deprivation and the ensuing reperfusion injury. METHODS Pertinent studies involving ischemic stroke, brain edema formation, astrocytes, and VP were identified by a search of the PubMed and the Web of Science databases in January 2016. Based on clinical findings and reports of animal experiments using ischemic stroke models, this systematic review reanalyzes the implication of individual reports in the edema formation and then establishes the inherent links among them. RESULTS This systematic review reveals that cytotoxic edema and vasogenic brain edema in classical view are mainly under the influence of a continuous malfunction of astrocytic plasticity. Adaptive VP secretion can modulate membrane ion transport, water permeability, and blood-brain barrier integrity, which are largely via changing astrocytic plasticity. Maladaptive VP hypersecretion leads to disruptions of ion and water balance across cell membranes as well as the integrity of the blood-brain barrier. This review highlights our current understandings of the cellular mechanisms underlying ischemic brain edema formation and its association with VP hypersecretion. CONCLUSIONS VP hypersecretion promotes brain edema formation in ischemic stroke by disrupting hydromineral balance in the neurovascular unit; suppressing VP hypersecretion has the potential to alleviate ischemic brain edema.
Collapse
Affiliation(s)
- Shu-Wei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiao-Yu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Stephani C Wang
- Department of Surgery, Albany Medical Center, Albany, New York
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| |
Collapse
|
44
|
Shi Y, Leak RK, Keep RF, Chen J. Translational Stroke Research on Blood-Brain Barrier Damage: Challenges, Perspectives, and Goals. Transl Stroke Res 2016; 7:89-92. [PMID: 26757714 PMCID: PMC4808584 DOI: 10.1007/s12975-016-0447-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 01/05/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Yejie Shi
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, S-507 Biomedical Science Tower 3500 Terrace Street, Pittsburgh, PA, 15213, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, S-507 Biomedical Science Tower 3500 Terrace Street, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
45
|
Changes in Brain Swelling and Infarction Volume over Four Days After Hypoxia Ischemia in Neonatal Rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:111-4. [PMID: 26463932 DOI: 10.1007/978-3-319-18497-5_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The leading cause of morbidity and mortality in infants is hypoxia-ischemia (HI). The current therapies for HI have limited success, in part due to a lack of understanding of HI pathophysiology and underlying mechanisms. Herein, a neonatal rat model of HI was used to examine the changes in brain swelling and infarct volume over 4 days after HI. Forty-four P10 rat pups were sacrificed at 2, 3, or 4 days post-HI. After sacrifice, the brains were removed, sliced, and stained with TTC (2,3,5-triphenyl-2H-tetrazolium chloride). Images of TTC-stained brains were used for measurement of the ipsilateral hemisphere brain volumes and infarct volumes, calculated using standard equations. The hemispheric brain volumes of HI animals in all groups was lower than that of sham animals and decreased as the post-HI sacrifice time increased. The infarct volume of HI animals was larger than that of sham animals. Infarct volumes tended to decrease over the days post-HI. The change in infarct volume is likely the result of a combination of brain growth and repair mechanisms. However, changes in the hemispheric brain volume may include tissue growth and repair mechanism, so also may be a limitation of the current algorithm used for calculating ipsilateral hemisphere brain volume.
Collapse
|
46
|
Methylene Blue Ameliorates Ischemia/Reperfusion-Induced Cerebral Edema: An MRI and Transmission Electron Microscope Study. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:227-36. [PMID: 26463954 DOI: 10.1007/978-3-319-18497-5_41] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The neuroprotective effect of methylene blue (MB) has been identified against various brain disorders, including ischemic stroke. In the present study, we evaluated the effects of MB on postischemic brain edema using magnetic resonance imaging (MRI) and transmission electron microscopy (TEM). Adult male rats were subjected to transient focal cerebral ischemia induced by 1 h middle cerebral artery occlusion (MCAO), followed by reperfusion. MB was infused intravenously immediately after reperfusion (3 mg/kg) and again at 3 h post-occlusion (1.5 mg/kg). Normal saline was administered as vehicle control. Sequential MRIs, including apparent diffusion coefficient (ADC) and T2-weighted imaging (T2WI), were obtained at 0.5, 2.5, and 48 h after the onset of stroke. Separated groups of animals were sacrificed at 2.5 and 48 h after stroke for ultrastructural analysis by TEM. In addition, final lesion volumes were analyzed by triphenyltetrazolium chloride (TTC) staining at 48 h after stroke. Ischemic stroke induced ADC lesion volume at 0.5 h during MCAOs that were temporally recovered at 1.5 h after reperfusion. No significant difference in ADC-defined lesion was observed between vehicle and MB treatment groups. At 48 h after stroke, MB significantly reduced ADC lesion and T2WI lesion volume and attenuated cerebral swelling. Consistently, MB treatment significantly decreased TTC-defined lesion volume at 48 h after stroke. TEM revealed remarkable swollen astrocytes, astrocytic perivascular end-feet, and concurrent shrunken neurons in the penumbra at 2.5 and 48 h after MCAO. MB treatment attenuated astrocyte swelling, the perivascular astrocytic foot process, and endothelium and also alleviated neuron degeneration. This study demonstrated that MB could decrease postischemic brain edema and provided additional evidence that future clinical investigation of MB for the treatment of ischemic stroke is warrented.
Collapse
|
47
|
Lekic T, Krafft PR, Klebe D, Flores J, Rolland WB, Tang J, Zhang JH. PAR-1, -4, and the mTOR Pathway Following Germinal Matrix Hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:213-6. [PMID: 26463951 DOI: 10.1007/978-3-319-18497-5_38] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Germinal matrix hemorrhage (GMH) is the most common cause of neurological complications of prematurity and has lasting implications. PAR-1 and PAR-4 receptors are involved with upstream signaling pathways following brain hemorrhage in adult models of stroke, of which the mammalian target of rapamycin (mTOR) is a potential downstream mediator. Therefore, we hypothesized a role for PAR-1, -4/ mTOR signaling following GMH brain injury. Postnatal day 7 Sprague-Dawley rats were subjected to GMH through stereotactic infusion of collagenase into the right ganglionic eminence. Rodents were euthanized at 72 h (short term), or 4 weeks (long term). Short-term mTOR expression was evaluated by Western blot in the context of PAR-1 (SCH-79797) and PAR-4 (P4pal10) inhibition. Pups in the long-term group were administered the selective mTOR inhibitor (rapamycin) with neurobehavioral and brain pathological examinations performed at 4 weeks. Pharmacological PAR-1, -4 antagonism normalized the increased mTOR expression following GMH. Early inhibition of mTOR by rapamycin improved long-term outcomes in rats. Mammalian-TOR signaling plays an important role in brain injury following neonatal GMH, possibly involving upstream PAR-1, -4 mechanisms.
Collapse
Affiliation(s)
- Tim Lekic
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Paul R Krafft
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Damon Klebe
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Jerry Flores
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - William B Rolland
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA. .,Department of Neurosurgery, School of Medicine, Loma Linda, CA, USA. .,Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall Rm 219, Loma Linda, CA, 92354, USA.
| |
Collapse
|
48
|
Osteopontin-Rac1 on Blood-Brain Barrier Stability Following Rodent Neonatal Hypoxia-Ischemia. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:263-7. [PMID: 26463959 DOI: 10.1007/978-3-319-18497-5_46] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Osteopontin (OPN) is a neuroprotective molecule that is upregulated following rodent neonatal hypoxic-ischemic (nHI) brain injury. Because Rac1 is a regulator of blood-brain barrier (BBB) stability, we hypothesized a role for this in OPN signaling. nHI was induced by unilateral ligation of the right carotid artery followed by hypoxia (8 % oxygen for 2 h) in P10 Sprague-Dawley rat pups. Intranasal (iN) OPN was administered at 1 h post-nHI. Groups consisted of: (1) Sham, (2) Vehicle, (3) OPN, and (4) OPN + Rac1 inhibitor (NSC23766). Evans blue dye extravasation (BBB permeability) was quantified 24 h post-nHI, and brain edema at 48 h. Increased BBB permeability and brain edema following nHI was ameliorated in the OPN treatment group. However, those rat pups receiving OPN co-treatment with the Rac1 inhibitor experienced no improvement compared with vehicle. OPN protects the BBB following nHI, and this was reversed by Rac1 inhibitor (NSC23766).
Collapse
|
49
|
Lekic T, Hardy M, Fujii M, McBride DW, Zhang JH. Brain Volume Determination in Subarachnoid Hemorrhage Using Rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:99-102. [PMID: 26463930 DOI: 10.1007/978-3-319-18497-5_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Brain edema is routinely measured using the wet-dry method. Volume, however, is the sum total of all cerebral tissues, including water. Therefore, volumetric change following injury may not be adequately quantified using percentage of edema. We thus tested the hypothesis that dried brains can be reconstituted with water and then re-measured to determine the actual volume. Subarachnoid hemorrhage (SAH) was induced by endovascular perforation in adult male Sprague-Dawley rats (n = 30). Animals were euthanized at 24 and 72 h after evaluation of neurobehavior for determination of brain water content. Dried brains were thereafter reconstituted with equal parts of water (lost from brain edema) and centrifuged to remove air bubbles. The total volume was quantified using hydrostatic (underwater) physics principles that 1 ml water (mass) = 1 cm(3) (volume). The amount of additional water needed to reach a preset level marked on 2-ml test tubes was added to that lost from brain edema, and from the brain itself, to determine the final volume. SAH significantly increased both brain water and volume while worsening neurological function in affected rats. Volumetric measurements demonstrated significant brain swelling after SAH, in addition to the brain edema approach. This modification of the "wet-dry" method permits brain volume determination using valuable post hoc dried brain tissue.
Collapse
Affiliation(s)
- Tim Lekic
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Maurice Hardy
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Mutsumi Fujii
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Devin W McBride
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Neurosurgery, School of Medicine, Loma Linda, CA, USA. .,Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall Rm 219, Loma Linda, CA, 92354, USA.
| |
Collapse
|
50
|
Abstract
Astrocyte endfeet envelop the cerebral capillaries that form the blood-brain barrier. Swelling of these endfeet occurs early in cerebral ischemia. It is generally hypothesized that such swelling occurs as the result of factors released from parenchymal brain cells during an ischemic stroke (e.g., K(+) and L-glutamate). In this review of mechanisms that can elicit astrocyte swelling in ischemic stroke, we hypothesize that, instead or in addition, such swelling may be a response to blood-brain barrier dysfunction. Astrocyte endfeet swelling may help form a cuff around a damaged vessel that limits the egress of plasma constituents and blood (hemorrhage) into brain.
Collapse
|