1
|
Qiao H, Wang S, Tao M, Fan H, Zhao T, Du Y, Dong M. Serum neurofilament heavy chain predicts post-stroke cognitive impairment. Sci Rep 2025; 15:13556. [PMID: 40253401 PMCID: PMC12009425 DOI: 10.1038/s41598-025-96952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
Serum phosphorylation neurofilament heavy chain (p-NfH) is a marker of axonal injury, and previous research has shown an association between p-NfH and both Alzheimer's disease and frontotemporal dementia. However, there have been no reports on its relationship with post-stroke cognitive impairment (PSCI). The purpose of this study is to investigate whether p-NfH can serve as a predictive biomarker for PSCI following acute ischemic stroke (AIS). From July 2020 to September 2021, a total of 58 cases of first-time acute ischemic stroke (AIS) patients were admitted to the Department of Neurology in the Second Hospital of Hebei Medical University. Additionally, 30 healthy volunteers were randomly selected as the control group. Demographic data, medical history, NIHSS scores, cerebral infarction volume, Fazekas scores for white matter and the serum p-NfH were collected. Follow-up assessments were conducted at 6 and 12 months after AIS. Cognitive function was evaluated using a multi-domain cognitive assessment scale, and patients were categorized into the post-stroke cognitive impairment group (PSCI) and non-post-stroke cognitive impairment group (N-PSCI). Further stratification was done into the progression group (MoCA score decline) and stable group (MoCA score unchanged or improved) based on the difference in MoCA scores between 12 and 6 months. The serum p-NfH levels in the AIS group were significantly higher than those in the control group (p < 0.01). Additionally, p-NfH levels were positively correlated with NIHSS scores and infarct volume. Furthermore, AIS patients with moderate to severe cerebral white matter lesions (Fazekas score ≥ 2) showed higher p-NfH levels compared to AIS patients with no or mild white matter lesions (Fazekas score 0 or 1) (p < 0.01). The PSCI group demonstrated higher p-NfH levels compared to the N-PSCI group, even after accounting for variables such as age, education level, NIHSS, infarct volume, and Fazekas grading (OR = 1.06, 95% CI 1.004-1.11, p = 0.03). Furthermore, the progression group exhibited significantly elevated p-NfH levels in comparison to the stable group. The ROC curve analysis revealed that the ideal cutoff point for p-NfH was determined to be at 166.03 pg/ml. This cutoff point exhibited a sensitivity of 0.774 and a specificity of 0.926 (p < 0.01). Furthermore, the area under the curve was calculated to be 0.881 (95% CI 0.791-0.97, p < 0.01). Serum p-NfH is a potential biomarker for predicting PSCI. Further investigation should explore its potential as an indicator for timely cognitive intervention in stroke patients during follow-up.
Collapse
Affiliation(s)
- Huimin Qiao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Suhuan Wang
- Department of Neurology, Xingtai Central Hospital, Xingtai, China
| | - Meichun Tao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Haolong Fan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Tianyi Zhao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Yuanyuan Du
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Mei Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
2
|
Karimkhani H, Shojaolsadati P, Yiğitbaşı T, Kolbası B, Emekli N. The effect of calpain inhibitor-I on copper oxide nanoparticle-induced damage and cerebral ischemia-reperfusion in a rat model. Biomed Pharmacother 2024; 174:116539. [PMID: 38615610 DOI: 10.1016/j.biopha.2024.116539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024] Open
Abstract
This study aimed to investigate the effects of the calpain inhibitor N-Acetyl-Leu-Leu-norleucinal (ALLN) on neuroapoptotic cell damage caused by Copper Oxide Nanoparticles (CuO-NP) and exacerbation of damage through brain ischemia/reperfusion (I/R) in a rat model. Male Wistar Albino rats (n=80) were divided into eight groups: Control, I/R, CuO-NP, CuO-NP+I/R, I/R+ALLN, CuO-NP+ALLN, CuO-NP+I/R+ALLN, and DMSO. Biochemical markers (MBP, S100B, NEFL, NSE, BCL-2, Cyt-C, Calpain, TNF-α, Caspase-3, MDA, and CAT) were measured in serum and brain tissue samples. Histological examinations (H&E staining), DNA fragmentation analysis (TUNEL) were performed, along with Caspase-3 assessment. The ALLN-treated groups exhibited significant improvements in biochemical markers and a remarkable reduction in apoptosis compared to the damaged groups (CuO-NP and I/R). H&E and Caspase-3 staining revealed damage-related morphological changes and reduced apoptosis in the ALLN-treated group. However, no differences were observed among the groups with TUNEL staining. The findings suggest that ALLN, as a calpain inhibitor, has potential implications for anti-apoptotic treatment, specifically in mitigating neuroapoptotic cell damage caused by CuO-NP and I/R.
Collapse
Affiliation(s)
- Hadi Karimkhani
- Department of Biochemistry, School of Medicine, Istanbul Okan University, Istanbul, Turkey; Department of Stem Cell, School of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey.
| | - Paria Shojaolsadati
- Department of Anatomy, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Türkan Yiğitbaşı
- Department of Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Bircan Kolbası
- Department of Histology and Embryology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Neslin Emekli
- Department of Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
3
|
Neuroprotective Effects of Nasopharyngeal Perfluorochemical Cooling in a Rat Model of Subarachnoid Hemorrhage. World Neurosurg 2018; 121:e481-e492. [PMID: 30267945 DOI: 10.1016/j.wneu.2018.09.142] [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: 02/13/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Subarachnoid hemorrhage (SAH) frequently results in severe morbidity, even mortality. Hypothermia is known to have a neuroprotective effect in ischemic injuries. The aim of this study was to determine whether nasopharyngeal (NP) perfluorochemical (PFC) cooling could be used in a rat model of SAH model for neuroprotection. METHODS SAH was induced in 16 male Sprague-Dawley rats by cisterna magna injection of 0.3 mL autologous blood. Vital signs, temperatures, cerebral blood flow (CBF), and brain histology were assessed. Brain cooling was performed on the treatment group using the NP-PFC method starting from 20 minutes after SAH. RESULTS No SAH-related deaths were observed in either group. SAH caused an immediate decrease in mean arterial pressure (17.0% ± 4.90% below baseline values). SAH induction caused a significant and rapid decrease in CBF from baseline (approximately -65%, ranging from -32% to -85%) in both hemispheres. In the left hemisphere, cooling facilitated the return of CBF to baseline values within 20 minutes of treatment with further increase in CBF that stabilized by the 2 hours after injury time point. Quantitative immunohistochemistry showed that there were significantly more NeuN-positive cells in the cortex and significantly fewer IBA-1-positive microglia and glial fibrillary acidic protein-positive astrocytes cells in both cortex and hippocampus in the animals that received NP-PFC cooling compared with no treatment, reflecting preserved neuronal integrity and reduced inflammation. CONCLUSIONS The data from this study indicate that local hypothermia by NP-PFC cooling supports return of CBF and neuronal integrity and suppresses the inflammatory response in SAH, suggestive of a promising neuroprotective approach in management of SAH.
Collapse
|
4
|
DeGracia DJ. Regulation of mRNA following brain ischemia and reperfusion. WILEY INTERDISCIPLINARY REVIEWS-RNA 2017; 8. [PMID: 28097803 DOI: 10.1002/wrna.1415] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/11/2016] [Accepted: 12/20/2016] [Indexed: 12/31/2022]
Abstract
There is growing appreciation that mRNA regulation plays important roles in disease and injury. mRNA regulation and ribonomics occur in brain ischemia and reperfusion (I/R) following stroke and cardiac arrest and resuscitation. It was recognized over 40 years ago that translation arrest (TA) accompanies brain I/R and is now recognized as part of the intrinsic stress responses triggered in neurons. However, neuron death correlates to a prolonged TA in cells fated to undergo delayed neuronal death (DND). Dysfunction of mRNA regulatory processes in cells fated to DND prevents them from translating stress-induced mRNAs such as heat shock proteins. The morphological and biochemical studies of mRNA regulation in postischemic neurons are discussed in the context of the large variety of molecular damage induced by ischemic injury. Open issues and areas of future investigation are highlighted. A sober look at the molecular complexity of ischemia-induced neuronal injury suggests that a network framework will assist in making sense of this complexity. The ribonomic network sits between the gene network and the various protein and metabolic networks. Thus, targeting the ribonomic network may prove more effective at neuroprotection than targeting specific molecular pathways, for which all efforts have failed to the present time to stop DND in stroke and after cardiac arrest. WIREs RNA 2017, 8:e1415. doi: 10.1002/wrna.1415 For further resources related to this article, please visit the WIREs website.
Collapse
|
5
|
Maniskas ME, Roberts JM, Trueman R, Learoyd AE, Gorman A, Fraser JF, Bix GJ. Intra-arterial nitroglycerin as directed acute treatment in experimental ischemic stroke. J Neurointerv Surg 2016; 10:29-33. [PMID: 28031354 DOI: 10.1136/neurintsurg-2016-012793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nitroglycerin (also known as glyceryl trinitrate (GTN)), a vasodilator best known for treatment of ischemic heart disease, has also been investigated for its potential therapeutic benefit in ischemic stroke. The completed Efficacy of Nitric Oxide in Stroke trial suggested that GTN has therapeutic benefit with acute (within 6 hours) transdermal systemic sustained release therapy. OBJECTIVE To examine an alternative use of GTN as an acute therapy for ischemic stroke following successful recanalization. METHODS We administered GTN IA following transient middle cerebral artery occlusion in mice. Because no standard dose of GTN is available following emergent large vessel occlusion, we performed a dose-response (3.12, 6.25, 12.5, and 25 µg/µL) analysis. Next, we looked at blood perfusion (flow) through the middle cerebral artery using laser Doppler flowmetry. Functional outcomes, including forced motor movement rotor rod, were assessed in the 3.12, 6.25, and 12.5 µg/µL groups. Histological analysis was performed using cresyl violet for infarct volume, and glial fibrillary activating protein (GFAP) and NeuN immunohistochemistry for astrocyte activation and mature neuron survival, respectively. RESULTS Overall, we found that acute post-stroke IA GTN had little effect on vessel dilatation after 15 min. Functional analysis showed a significant difference between GTN (3.12 and 6.25 µg/µL) and control at post-stroke day 1. Histological measures showed a significant reduction in infarct volume and GFAP immunoreactivity and a significant increase in NeuN. CONCLUSIONS These results demonstrate that acute IA GTN is neuroprotective in experimental ischemic stroke and warrants further study as a potentially new stroke therapy.
Collapse
Affiliation(s)
- Michael E Maniskas
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, Kentucky, USA.,Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA.,Department of Neurosurgery, University of Kentucky, Lexington, Kentucky, USA
| | - Jill M Roberts
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, Kentucky, USA.,Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Rebecca Trueman
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Amanda Gorman
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Justin F Fraser
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, Kentucky, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA.,Department of Neurosurgery, University of Kentucky, Lexington, Kentucky, USA.,Department of Neurology, University of Kentucky, Lexington, Kentucky, USA.,Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Gregory J Bix
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, Kentucky, USA.,Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA.,Department of Neurosurgery, University of Kentucky, Lexington, Kentucky, USA.,Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
6
|
Up-regulation of neurofilament light chains is associated with diminished immunoreactivities for MAP2 and tau after ischemic stroke in rodents and in a human case. J Chem Neuroanat 2016; 78:140-148. [DOI: 10.1016/j.jchemneu.2016.09.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 11/18/2022]
|
7
|
Jiang MQ, Zhao YY, Cao W, Wei ZZ, Gu X, Wei L, Yu SP. Long-term survival and regeneration of neuronal and vasculature cells inside the core region after ischemic stroke in adult mice. Brain Pathol 2016; 27:480-498. [PMID: 27514013 DOI: 10.1111/bpa.12425] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 07/27/2016] [Indexed: 12/20/2022] Open
Abstract
Focal cerebral ischemia results in an ischemic core surrounded by the peri-infarct region (penumbra). Most research attention has been focused on penumbra while the pattern of cell fates inside the ischemic core is poorly defined. In the present investigation, we tested the hypothesis that, inside the ischemic core, some neuronal and vascular cells could survive the initial ischemic insult while regenerative niches might exist many days after stroke in the adult brain. Adult mice were subjected to focal cerebral ischemia induced by permanent occlusion of distal branches of the middle cerebral artery (MCA) plus transient ligations of bilateral common carotid artery (CCA). The ischemic insult uniformly reduced the local cerebral blood flow (LCBF) by 90%. Massive cell death occurred due to multiple mechanisms and a significant infarction was cultivated in the ischemic cortex 24 h later. Nevertheless, normal or even higher levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) persistently remained in the core tissue, some NeuN-positive and Glut-1/College IV-positive cells with intact ultrastructural features resided in the core 7-14 days post stroke. BrdU-positive but TUNEL-negative neuronal and endothelial cells were detected in the core where extensive extracellular matrix infrastructure developed. Meanwhile, GFAP-positive astrocytes accumulated in the penumbra and Iba-1-positive microglial/macrophages invaded the core several days after stroke. The long term survival of neuronal and vascular cells inside the ischemic core was also seen after a severe ischemic stroke induced by permanent embolic occlusion of the MCA. We demonstrate that a therapeutic intervention of pharmacological hypothermia could save neurons/endothelial cells inside the core. These data suggest that the ischemic core is an actively regulated brain region with residual and newly formed viable neuronal and vascular cells acutely and chronically after at least some types of ischemic strokes.
Collapse
Affiliation(s)
- Michael Qize Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA
| | - Ying-Ying Zhao
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA.,Department of Neurology, Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenyuan Cao
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA.,Department of Neurology, Friendship Hospital, Capital Medical University, Beijing, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA
| |
Collapse
|
8
|
Maniskas ME, Roberts JM, Aron I, Fraser JF, Bix GJ. Stroke neuroprotection revisited: Intra-arterial verapamil is profoundly neuroprotective in experimental acute ischemic stroke. J Cereb Blood Flow Metab 2016; 36:721-30. [PMID: 26661189 PMCID: PMC4821022 DOI: 10.1177/0271678x15608395] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/21/2015] [Indexed: 12/20/2022]
Abstract
While clinical trials have now solidified the role of thrombectomy in emergent large vessel occlusive stroke, additional therapies are needed to optimize patient outcome. Using our previously described experimental ischemic stroke model for evaluating adjunctive intra-arterial drug therapy after vessel recanalization, we studied the potential neuroprotective effects of verapamil. A calcium channel blocker, verapamil is often infused intra-arterially by neurointerventionalists to treat cerebral vasospasm. Such a direct route of administration allows for both focused targeting of stroke-impacted brain tissue and minimizes potential systemic side effects. Intra-arterial administration of verapamil at a flow rate of 2.5 µl/min and injection volume of 10 µl immediately after middle cerebral artery recanalization in C57/Bl6 mice was shown to be profoundly neuroprotective as compared to intra-arterial vehicle-treated stroke controls. Specifically, we noted a significant (P ≤ 0.05) decrease in infarct volume, astrogliosis, and cellular apoptosis as well as a significant increase in neuronal survival and functional outcome over seven days. Furthermore, intra-arterial administration of verapamil was well tolerated with no hemorrhage, systemic side effects, or increased mortality. Thus, verapamil administered intra-arterially immediately following recanalization in experimental ischemic stroke is both safe and neuroprotective and merits further study as a potential therapeutic adjunct to thrombectomy.
Collapse
Affiliation(s)
- Michael E Maniskas
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, USA Sanders Brown Center on Aging, University of Kentucky, Lexington, USA
| | - Jill M Roberts
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, USA Sanders Brown Center on Aging, University of Kentucky, Lexington, USA
| | - Ishi Aron
- Sanders Brown Center on Aging, University of Kentucky, Lexington, USA
| | - Justin F Fraser
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, USA Department of Neurosurgery, University of Kentucky, Lexington, USA Department of Neurology, University of Kentucky, Lexington, Kentucky Department of Radiology, University of Kentucky, Lexington, USA
| | - Gregory J Bix
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, USA Sanders Brown Center on Aging, University of Kentucky, Lexington, USA Department of Neurology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
9
|
A review on animal models of stroke: An update. Brain Res Bull 2016; 122:35-44. [PMID: 26902651 DOI: 10.1016/j.brainresbull.2016.02.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 01/11/2023]
Abstract
Stroke is one of the major healthcare challenges prevailing across the globe due to its significant rate of mortality and morbidity. Stroke is multifactorial in nature and involves several cellular and molecular signaling cascades that make the pathogenesis complex and treatment difficult. For a deeper understanding of the diverse pathological mechanisms and molecular & cellular cascades during stroke, animal modeling serves as a reliable and an effective tool. This also helps to develop and critically analyse various neuroprotective strategies for the mitigation of this devastating disease. Animal modeling for stroke has been revolutionized with the development of newer and more relevant models or approaches that mimic the clinical setting of stroke to a greater extent. This review analyses experimental models of stroke (ischemic and hemorrhagic) and their reliability in stroke situation. Besides this, the review also stresses upon the use of various preclinical models to understand the pathophysiological mechanisms that operate during stroke and to elucidate new, safe and effective neuroprotective agents to combat this life threatening healthcare concern.
Collapse
|
10
|
Abstract
UNLABELLED After ischemic stroke, various damage-associated molecules are released from the ischemic core and diffuse to the ischemic penumbra, activating microglia and promoting proinflammatory responses that may cause damage to the local tissue. Here we demonstrate using in vivo and in vitro models that, during sublethal ischemia, local neurons rapidly produce interleukin-4 (IL-4), a cytokine with potent anti-inflammatory properties. One such anti-inflammatory property includes its ability to polarize macrophages away from a proinflammatory M1 phenotype to a "healing" M2 phenotype. Using an IL-4 reporter mouse, we demonstrated that IL-4 expression was induced preferentially in neurons in the ischemic penumbra but not in the ischemic core or in brain regions that were spared from ischemia. When added to cultured microglia, IL-4 was able to induce expression of genes typifying the M2 phenotype and peroxisome proliferator activated receptor γ (PPARγ) activation. IL-4 also enhanced expression of the IL-4 receptor on microglia, facilitating a "feedforward" increase in (1) their expression of trophic factors and (2) PPARγ-dependent phagocytosis of apoptotic neurons. Parenteral administration of IL-4 resulted in augmented brain expression of M2- and PPARγ-related genes. Furthermore, IL-4 and PPARγ agonist administration improved functional recovery in a clinically relevant mouse stroke model, even if administered 24 h after the onset of ischemia. We propose that IL-4 is secreted by ischemic neurons as an endogenous defense mechanism, playing a vital role in the regulation of brain cleanup and repair after stroke. Modulation of IL-4 and its associated pathways could represent a potential target for ischemic stroke treatment. SIGNIFICANCE STATEMENT Depending on the activation signal, microglia/macrophages (MΦ) can behave as "healing" (M2) or "harmful" (M1). In response to ischemia, damaged/necrotic brain cells discharge factors that polarize MΦ to a M1-like phenotype. This polarization emerges early after stroke and persists for days to weeks, driving secondary brain injury via proinflammatory mediators and oxidative damage. Our study demonstrates that, to offset this M1-like polarization process, sublethally ischemic neurons may instead secrete a potent M2 polarizing cytokine, interleukin-4 (IL-4). In the presence of IL-4 (including when IL-4 is administered exogenously), MΦ become more effective in the cleanup of ischemic debris and produce trophic factors that may promote brain repair. We propose that IL-4 could represent a potential target for ischemic stroke treatment/recovery.
Collapse
|
11
|
McCombe PA, Pfluger C, Singh P, Lim CYH, Airey C, Henderson RD. Serial measurements of phosphorylated neurofilament-heavy in the serum of subjects with amyotrophic lateral sclerosis. J Neurol Sci 2015; 353:122-9. [PMID: 25958264 DOI: 10.1016/j.jns.2015.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 12/31/2022]
Abstract
There is a need for a blood biomarker of disease activity in ALS. This marker needs to measure the loss of motor neurones. Phosphorylated neurofilament heavy chain (pNfH) in the serum is a biomarker of axonal injury. Previous studies have found that levels of pNfH are elevated in ALS. We have performed a serial study of pNfH levels in 98 subjects from our ALS clinic. There was significant elevation of levels of pNfH in subjects with ALS compared to controls, although there was considerable variability. In studies of individuals who had two or more serial samples, we found that the levels of pNfH increased over time in the early stage of disease. Levels were low in subjects with long survival. The rate of rise of pNfH was inversely correlated with survival. We suggest that the initial level of pNfH is a marker of disease severity and that changes in pNfH levels are markers of disease progression.
Collapse
Affiliation(s)
- P A McCombe
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia.
| | - C Pfluger
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - P Singh
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - C Y H Lim
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - C Airey
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - R D Henderson
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| |
Collapse
|
12
|
Mashita T, Kamishina H, Nakamoto Y, Akagi Y, Nakanishi A, Harasaki Y, Ozawa T, Uemura T, Kobatake Y, Shimamura S, Kitamura N, Maeda S, Uzuka Y, Shaw G, Yasuda J. Combination of serum phosphorylated neurofilament heavy subunit and hyperintensity of intramedullary T2W on magnetic resonance imaging provides better prognostic value of canine thoracolumbar intervertebral disc herniation. J Vet Med Sci 2015; 77:433-8. [PMID: 25650056 PMCID: PMC4427744 DOI: 10.1292/jvms.14-0582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to evaluate the prognostic value of concurrent measurement of serum phosphorylated neurofilament heavy subunit (pNF-H) concentration and intramedullary T2W hyperintensity in paraplegic to paraplegic dogs. Our hypothesis was that concurrent measurement of these would provide a more accurate prediction of functional outcome in dogs with thoracolumbar intervertebral disc herniation (IVDH). A prospective case-control clinical study was designed using 94 dogs with acute onset of thoracolumbar IVDH. The association of serum pNF-H concentration, T2W hyperintensity on sagittal MRI (T2H/L2), deep pain perception and surgical outcome were evaluated with logistic regression analysis after three months for all 94 surgically treated dogs. Sensitivity to predict non-ambulatory outcome was compared among pNF-H and T2H/L2 and combination of both. Logistic regression analysis indicated that serum pNF-H concentration and T2H/L2 were significantly correlated with surgical outcome (P<0.05); however, deep pain perception was not (P=0.41). The results of logistic regression analysis indicated that the odds ratios of unsuccessful long-term outcome were 2.6 for serum pNF-H concentration, 1.9 for T2H/L2 and 2.3 for deep pain sensation. The sensitivity and specificity to predict non-ambulatory outcome for using serum parameter pNF-H>2.6 ng/ml, using T2H/L2 value of>0.84 and using both serum pNF-H and T2H/L2, were 95% and 75.7%, 65% and 86.5%, and 90.0% and 97.5%, respectively. Therefore, combined measurements of serum pNF-H and T2H/L2 might be useful for predicting long-term outcome in dogs with thoracolumbar IVDH.
Collapse
Affiliation(s)
- Tadahisa Mashita
- Maizuru Animal Medical Center, 8-22 Hamamachi, Kyoto; Department of Clinical Veterinary Science, United Graduate School of Veterinary Science, Gifu University, 1-1 Yanagito 509-1193, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Maniskas M, Bix G, Fraser J. Selective intra-arterial drug administration in a model of large vessel ischemia. J Neurosci Methods 2014; 240:22-7. [PMID: 25445249 DOI: 10.1016/j.jneumeth.2014.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 10/24/2022]
Abstract
With continuing disconnect between laboratory stroke treatment models and clinical stroke therapy, we propose a novel experimental model to study stroke and vessel recanalization that mirrors acute management of large vessel stroke, with concomitant directed pharmacotherapy. Using the tandem transient ipsilateral common carotid/middle cerebral artery occlusion (MCAO) model to induce stroke in mice we then added selective intra-arterial (IA) drug administration for directed pharmacotherapy. The IA model uses micro-angio tubing placed at the bifurcation of the CCA to selectively administer the drug to the internal carotid distribution. We have shown that delivery of pharmacotherapy agents selectively through an IA injection is feasible in a mouse model, which will permit studies involving pharmacotherapy, transgenic modification, and/or a combination. Our IA model has similarities to previously published models of IA injection but differs in that we do not leave an indwelling micro-port or catheter in our animals, which is not clinically relevant as it does not reflect the human condition or current clinical management. Furthermore, we optimized our model to selectively direct therapy to the ipsilateral, stroke affected hemisphere. By developing an IA drug delivery model that mirrors clinical conditions, we are bridging the gap between basic stroke research and what is standard practice in acute ischemic stroke intervention. The IA model of drug delivery can target agents directly to the site of injury while blunting systemic effects, dose penetration issues, and administration delay that have plagued the intraperitoneal and oral drug administration models.
Collapse
Affiliation(s)
- Michael Maniskas
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY, United States; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Gregory Bix
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY, United States; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States; Department of Neurology, University of Kentucky, Lexington, KY, United States.
| | - Justin Fraser
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY, United States; Department of Neurosurgery, University of Kentucky, Lexington, KY, United States; Department of Neurology, University of Kentucky, Lexington, KY, United States; Department of Radiology, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
14
|
Predictive value of phosphorylated axonal neurofilament subunit H for clinical outcome in patients with acute intracerebral hemorrhage. Clin Chim Acta 2013; 424:182-6. [DOI: 10.1016/j.cca.2013.06.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/15/2013] [Accepted: 06/17/2013] [Indexed: 11/21/2022]
|
15
|
Gaur V, Kumar A. Effect of nonselective and selective COX-2 inhibitors on memory dysfunction, glutathione system, and tumor necrosis factor alpha level against cerebral ischemia reperfusion injury. Drug Chem Toxicol 2011; 35:218-24. [DOI: 10.3109/01480545.2011.589850] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
16
|
Moreira MS, Velasco IT, Ferreira LS, Ariga SKK, Abatepaulo F, Grinberg LT, Marques MM. Effect of laser phototherapy on wound healing following cerebral ischemia by cryogenic injury. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2011; 105:207-15. [PMID: 22024356 DOI: 10.1016/j.jphotobiol.2011.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 08/09/2011] [Accepted: 09/28/2011] [Indexed: 11/29/2022]
Abstract
Laser phototherapy emerges as an alternative or auxiliary therapy for acute ischemic stroke, traumatic brain injury, degenerative brain disease, spinal cord injury, and peripheral nerve regeneration, but its effects are still controversial. We have previously found that laser phototherapy immunomodulates the response to focal brain damage. Following direct cortical cryogenic injury the effects of laser phototherapy on inflammation and repair was assessed after cryogenic injury (CI) to the central nervous system (CNS) of rats. The laser phototherapy was carried out with a 780 nm AlGaAs diode laser. The irradiation parameters were: power of 40 mW, beam area of 0.04 cm(2), energy density of 3 J/cm(2) (3s) in two points (0.12 J per point). Two irradiations were performed at 3 h-intervals, in contact mode. Rats (20 non-irradiated - controls and 20 irradiated) were used. The wound healing in the CNS was followed in 6 h, 1, 7 and 14 days after the last irradiation. The size of the lesions, the neuron cell viability percentages and the amount of positive GFAP labeling were statistically compared by ANOVA complemented by Tukey's test (p<0.05). The distribution of lymphocytes, leukocytes and macrophages were also analyzed. CI created focal lesions in the cortex represented by necrosis, edema, hemorrhage and inflammatory infiltrate. The most striking findings were: lased lesions showed smaller tissue loss than control lesions in 6 h. During the first 24 h the amount of viable neurons was significantly higher in the lased group. There was a remarkable increase in the amount of GFAP in the control group by 14 days. Moreover, the lesions of irradiated animals had fewer leukocytes and lymphocytes in the first 24 h than controls. Considering the experimental conditions of this study it was concluded that laser phototherapy exerts its effect in wound healing following CI by controlling the brain damage, preventing neuron death and severe astrogliosis that could indicate the possibility of a better clinical outcome.
Collapse
Affiliation(s)
- Maria S Moreira
- School of Dentistry, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | | | | | | | | | | | | |
Collapse
|
17
|
Lee B, Clarke D, Al Ahmad A, Kahle M, Parham C, Auckland L, Shaw C, Fidanboylu M, Orr AW, Ogunshola O, Fertala A, Thomas SA, Bix GJ. Perlecan domain V is neuroprotective and proangiogenic following ischemic stroke in rodents. J Clin Invest 2011; 121:3005-23. [PMID: 21747167 DOI: 10.1172/jci46358] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 05/18/2011] [Indexed: 12/27/2022] Open
Abstract
Stroke is the leading cause of long-term disability and the third leading cause of death in the United States. While most research thus far has focused on acute stroke treatment and neuroprotection, the exploitation of endogenous brain self-repair mechanisms may also yield therapeutic strategies. Here, we describe a distinct type of stroke treatment, the naturally occurring extracellular matrix fragment of perlecan, domain V, which we found had neuroprotective properties and enhanced post-stroke angiogenesis, a key component of brain repair, in rodent models of stroke. In both rat and mouse models, Western blot analysis revealed elevated levels of perlecan domain V. When systemically administered 24 hours after stroke, domain V was well tolerated, reached infarct and peri-infarct brain vasculature, and restored stroke-affected motor function to baseline pre-stroke levels in these multiple stroke models in both mice and rats. Post-stroke domain V administration increased VEGF levels via a mechanism involving brain endothelial cell α5β1 integrin, and the subsequent neuroprotective and angiogenic actions of domain V were in turn mediated via VEGFR. These results suggest that perlecan domain V represents a promising approach for stroke treatment.
Collapse
Affiliation(s)
- Boyeon Lee
- Department of Molecular and Cellular Medicine, Texas A&M College of Medicine, College Station, Texas 77843, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Singh P, Yan J, Hull R, Read S, O'Sullivan J, Henderson RD, Rose S, Greer JM, McCombe PA. Levels of phosphorylated axonal neurofilament subunit H (pNfH) are increased in acute ischemic stroke. J Neurol Sci 2011; 304:117-21. [PMID: 21349546 DOI: 10.1016/j.jns.2011.01.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 12/29/2010] [Accepted: 01/27/2011] [Indexed: 12/31/2022]
Abstract
For the study of stroke outcomes, there is the need for measurements of severity of stroke damage. Phosphorylated neurofilament heavy protein (pNfH) levels are elevated in axonal injury. We have measured levels of pNfH in stroke and correlated these levels with measures of stroke severity. Blood samples were collected from 54 ischaemic stroke patients at day 1, week 1 (days 7-10) and weeks 3-6, and an ELISA was used to measure pNfH levels in each patient at each time-point. Serum pNfH levels were significantly elevated in stroke patients compared to healthy controls. The levels were low at day 1, higher at day 7 and reached a peak at week 3, the latest day that we assessed. Significant associations were found between the pNfH levels at week 3 and early and stroke severity, size and outcome. Blood pNfH levels that reflect the severity of ischaemic stroke, are correlated with outcome and rise during the weeks after stroke. This may be a useful measure of tissue damage in stroke.
Collapse
Affiliation(s)
- P Singh
- The University of Queensland, UQ Centre for Clinical Research, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhao X, Strong R, Piriyawat P, Palusinski R, Grotta JC, Aronowski J. Caffeinol at the receptor level: anti-ischemic effect of N-methyl-D-aspartate receptor blockade is potentiated by caffeine. Stroke 2009; 41:363-7. [PMID: 20044532 DOI: 10.1161/strokeaha.109.562900] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE Although caffeinol (a combination of a low dose of caffeine and ethanol) was shown to robustly reduce stroke damage in experimental models and is now in clinical evaluation for treatment of ischemic stroke, little is known about the potential mechanism of its action. METHODS We used an in vivo excitotoxicity model based on intracortical infusion of N-methyl-D-aspartate (NMDA) and a model of reversible focal ischemia to demonstrate NMDA receptor inhibition as a potential mechanism of caffeinol anti-ischemic activity. RESULTS Caffeinol reduced the size of excitotoxic lesion, and substitution of ethanol in caffeinol with the NMDA antagonists CNS-1102 and MK-801 but not with MgSO(4) produced treatment with strong synergistic effect that was at least as robust in reducing ischemic damage as caffeinol. This NMDA receptor antagonist and caffeine combination demonstrated a long window of opportunity, activity in spontaneously hypertensive rats, and, unlike caffeinol, was fully effective in animals chronically pretreated with ethanol. CONCLUSIONS Our study suggests that antiexcitotoxic properties may underlie some of the anti-ischemic effect of caffeinol. This study provides strong evidence that the anti-ischemic effect of NMDA receptor blockers in general can be dramatically augmented by caffeine, thus opening a possibility for new use of NMDA-based pharmacology in the treatment of stroke.
Collapse
Affiliation(s)
- Xiurong Zhao
- Stroke Program, Department of Neurology, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
20
|
Neuronal PPARgamma deficiency increases susceptibility to brain damage after cerebral ischemia. J Neurosci 2009; 29:6186-95. [PMID: 19439596 DOI: 10.1523/jneurosci.5857-08.2009] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) plays a role in regulating a myriad of biological processes in virtually all brain cell types, including neurons. We and others have reported recently that drugs which activate PPARgamma are effective in reducing damage to brain in distinct models of brain disease, including ischemia. However, the cell type responsible for PPARgamma-mediated protection has not been established. In response to ischemia, PPARgamma gene is robustly upregulated in neurons, suggesting that neuronal PPARgamma may be a primary target for PPARgamma-agonist-mediated neuroprotection. To understand the contribution of neuronal PPARgamma to ischemic injury, we generated conditional neuron-specific PPARgamma knock-out mice (N-PPARgamma-KO). These mice are viable and appeared to be normal with respect to their gross behavior and brain anatomy. However, neuronal PPARgamma deficiency caused these mice to experience significantly more brain damage and oxidative stress in response to middle cerebral artery occlusion. The primary cortical neurons harvested from N-PPARgamma-KO mice, but not astroglia, exposed to ischemia in vitro demonstrated more damage and a reduced expression of numerous key gene products that could explain increased vulnerability, including SOD1 (superoxide dismutase 1), catalase, glutathione S-transferase, uncoupling protein-1, or transcription factor liver X receptor-alpha. Also, PPARgamma agonist-based neuroprotective effect was lost in neurons from N-PPARgamma neurons. Therefore, we conclude that PPARgamma in neurons play an essential protective function and that PPARgamma agonists may have utility in neuronal self-defense, in addition to their well established anti-inflammatory effect.
Collapse
|
21
|
Diaz-Ruiz A, Zavala C, Montes S, Ortiz-Plata A, Salgado-Ceballos H, Orozco-Suarez S, Nava-Ruiz C, Pérez-Neri I, Perez-Severiano F, Ríos C. Antioxidant, antiinflammatory and antiapoptotic effects of dapsone in a model of brain ischemia/reperfusion in rats. J Neurosci Res 2009; 86:3410-9. [PMID: 18615706 DOI: 10.1002/jnr.21775] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Although dapsone (4,4'-diaminodiphenylsulfone) has been described as a neuroprotective agent in occlusive focal ischemia in rats, its mechanism of action is still unknown. To explore this mechanism, oxidative, inflammatory and apoptotic processes were evaluated in the striatum of adult rats using a model of ischemia-reperfusion (I/R), either with or without dapsone treatment. Male Wistar rats were submitted to transient middle cerebral artery occlusion for 2 hr, followed by reperfusion. Rats were dosed either with dapsone (12.5 mg/kg i.p.) or vehicle 30 min before or 30 min after the ischemia onset. Lipid peroxidation (LP) and nitrotyrosine contents were measured 22 hr after reperfusion, and myeloperoxidase activity was evaluated 46 hr after I/R. Different markers for apoptosis and necrosis were also evaluated both at 24 and 72 hr after I/R experimental procedure. LP increased by 37% in ischemic animals vs controls, and this effect was reversed by dapsone treatments. A similar effect was observed regarding nitrotyrosine striatal contents. Myeloperoxidase activity, a marker of inflammatory response, increased 3.7-fold in ischemic animals vs. control rats, and dapsone treatment antagonized that effect. Although apoptosis was increased by the effect of ischemia at both evaluation times, dapsone antagonized that effect only at 72 hr after surgery. Dapsone antagonized all of the I/R end points measured, showing a remarkable ability to decrease markers of damage through antioxidant, antiinflammatory, and anti-apoptotic effects.
Collapse
Affiliation(s)
- Araceli Diaz-Ruiz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez S.S.A., D.F. México, México
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hashimoto M, Zhao L, Nowak TS. Temporal thresholds for infarction and hypothermic protection in Long-Evans rats: factors affecting apparent 'reperfusion injury' after transient focal ischemia. Stroke 2008; 39:421-6. [PMID: 18174482 DOI: 10.1161/strokeaha.107.495788] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Some previous studies in Long-Evans rats noted larger infarcts after transient middle cerebral artery (MCA) occlusions than after permanent occlusions, interpreted to demonstrate "reperfusion injury." Recent experiments failed to reproduce this phenomenon, prompting an investigation of the sources of variability in this animal model. METHODS Male Long-Evans rats were subjected to surgical occlusion of the right MCA and ipsilateral common carotid artery. Variables tested included duration of occlusion and halothane anesthesia exposure and targeting of proximal or distal MCA occlusion sites. The temporal window for hypothermic protection was also investigated. RESULTS MCA occlusions at the level of the rhinal fissure produced graded increases in infarct volume with ischemia duration, and lesion size did not differ between 3-hour and permanent occlusions independent of anesthesia duration. Occlusions at a more distal site produced infarcts of comparable size after transient 3-hour occlusions and after permanent occlusions accompanied by prolonged anesthesia, but significantly smaller infarcts were seen when permanent occlusions were followed by rapid anesthesia termination. Hypothermia conferred protection only when initiated before reperfusion after transient proximal occlusions. CONCLUSIONS These results indicate that previously described "reperfusion injury" after transient MCA occlusions conversely reflects unexpected injury reduction when rats with permanent occlusions experience early anesthesia termination. More rapid blood pressure recovery under such conditions permits improved collateral perfusion. The absence of a detectable postischemic window for hypothermic protection further argues against a significant component of delayed postreperfusion injury in this model.
Collapse
Affiliation(s)
- Megumi Hashimoto
- Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Ave, Link 415, Memphis, TN 38163, USA
| | | | | |
Collapse
|
23
|
Yap E, Tan WL, Ng I, Ng YK. Combinatorial-approached neuroprotection using pan-caspase inhibitor and poly (ADP-ribose) polymerase (PARP) inhibitor following experimental stroke in rats; is there additional benefit? Brain Res 2007; 1195:130-8. [PMID: 18207135 DOI: 10.1016/j.brainres.2007.12.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Revised: 12/14/2007] [Accepted: 12/14/2007] [Indexed: 01/01/2023]
Abstract
Energy requiring apoptosis and presumably unregulated necrosis are considered conceptually and morphologically distinct forms of cell death which have been initially identified as two exclusive pathways. However, several apoptotic characteristics have been observed in the necrotic core lesion in ischemia which led to the controversial theory that cell death advances via a number of hybrid pathways among a continuum between the two processes. ATP availability has been shown to influence the decision between apoptosis and necrosis. The aims of our study are 1) to determine if combined inhibitors administration of pan-caspase inhibitor Carbobenzoxy-Val-Ala-Asp-fluoromethylketone (z-VAD-fmk) and non-selective poly (ADP-ribose) polymerase (PARP) inhibitor 3-aminobenzamide (3-AB) can further reduce infarct volume compared to single modality of either inhibitor following ischemic insult, 2) to ascertain the pharmacological intervention up to 24 hour post-middle cerebral artery occlusion (MCAo), and 3) to correlate intracellular ATP level with infarct volume. Single modality treatment was optimised at 3 mg/kg z-VAD-fmk and 30 mg/kg 3-AB with infarct volume measured at 24.13%+/-3.89% and 26.98%+/-2.22% respectively, while untreated control group was determined at 45.97%+/-1.86%. Combined inhibitors treatment rendered further reduction in infarct volume, measuring 7.228%+/-1.988%, 21.02%+/-1.06%, 24.40%+/-2.12% at 30 min, 6 h, 24 h post-ischemia respectively. In conclusion, the combined inhibitors administration of both z-VAD-fmk and 3-AB show further increased in infarct volume reduction with our ischemic model up to the 24 hour post-MCAo. However, in our in vivo study, no correlation between intracellular ATP level and infarct size was established.
Collapse
Affiliation(s)
- Elgin Yap
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, MD10, 4 Medical Drive, Singapore 117597
| | | | | | | |
Collapse
|
24
|
Zhao X, Sun G, Zhang J, Strong R, Song W, Gonzales N, Grotta JC, Aronowski J. Hematoma resolution as a target for intracerebral hemorrhage treatment: Role for peroxisome proliferator-activated receptor γ in microglia/macrophages. Ann Neurol 2007; 61:352-62. [PMID: 17457822 DOI: 10.1002/ana.21097] [Citation(s) in RCA: 298] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Phagocytosis is necessary to eliminate the hematoma after intracerebral hemorrhage (ICH); however, release of proinflammatory mediators and free radicals during phagocyte activation is toxic to neighboring cells, leading to secondary brain injury. Promotion of phagocytosis in a timely and efficient manner may limit the toxic effects of persistent blood products on surrounding tissue and may be important for recovery after ICH. METHODS Intrastriatal blood injection in rodents and primary microglia in culture exposed to red blood cells were used to model ICH and to study mechanisms of hematoma resolution and phagocytosis regulation by peroxisome proliferator-activated receptor gamma (PPARgamma) in microglia/macrophages. RESULTS Our study demonstrated that the PPARgamma agonist, rosiglitazone, promoted hematoma resolution, decreased neuronal damage, and improved functional recovery in a mouse ICH model. Microglia isolated from murine brains showed more efficient phagocytosis in response to PPARgamma activators. PPARgamma activators significantly increased PPARgamma-regulated gene (catalase and CD36) expression, whereas reducing proinflammatory gene (tumor necrosis factor-alpha, interleukin-1beta, matrix metalloproteinase-9, and inducible nitric oxide synthase) expression, extracellular H(2)O(2) level, and neuronal damage. Phagocytosis by microglia was significantly inhibited by PPARgamma gene knockdown or neutralizing anti-CD36 antibody, whereas it was enhanced by exogenous catalase. INTERPRETATION PPARgamma in macrophages acts as an important factor in promoting hematoma absorption and protecting other brain cells from ICH-induced damage.
Collapse
Affiliation(s)
- Xiurong Zhao
- Stroke Program, Department of Neurology, University of Texas-Houston Medical School, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhao X, Zhang Y, Strong R, Zhang J, Grotta JC, Aronowski J. Distinct patterns of intracerebral hemorrhage-induced alterations in NF-κB subunit, iNOS, and COX-2 expression. J Neurochem 2006; 101:652-63. [PMID: 17250675 DOI: 10.1111/j.1471-4159.2006.04414.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Transcription factor nuclear factor-kappaB (NF-kappaB), plays a key role in regulating inflammation in brain pathologies. The goal of this study was to characterize temporal changes in NF-kappaB activation, NF-kappaB subunit expression, and expression of selected NF-kappaB-regulated gene products [inducible form of nitric oxide synthase (iNOS) and cyclooxygenase-2], at the transcriptional and translational level in the brain after intracerebral hemorrhage (ICH). Employing the intrastriatal injection of autologous blood in rats to model ICH, we demonstrated, using NF-kappaB-DNA binding assay, a robust and prolonged NF-kappaB activation, starting as early as 15 min after the onset of ICH. Consequently, we demonstrated that the mRNA and protein for p50, p52, p65, c-Rel, and RelB NF-kappaB subunits, as well as IkappaBalpha were all up-regulated, with a time course ranging from minutes to days following ICH, depending on the subunit. Using reverse transcription-polymerase chain reaction to analyze mRNA and immunoblotting to analyze protein in ICH-affected tissue, we found robust induction of iNOS at both mRNA and protein levels that followed a time-course similar to changes in p65, p52, and RelB mRNA. Oddly, in contrast to iNOS, cyclooxygenase-2 mRNA and protein following an early transient increase demonstrated significant reduction in response to ICH. In summary, NF-kappaB activation occurs within minutes and persists for at least a week in response to ICH. This reaction utilizes different NF-kappaB regulatory subunits and is associated with the expression of selected target genes.
Collapse
Affiliation(s)
- Xiurong Zhao
- Department of Neurology, Stroke Program, University of Texas-Houston Medical School, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
26
|
Ou Z, Zhao X, Labiche LA, Strong R, Grotta JC, Herrmann O, Aronowski J. Neuronal expression of peroxisome proliferator-activated receptor-gamma (PPARγ) and 15d-prostaglandin J2—Mediated protection of brain after experimental cerebral ischemia in rat. Brain Res 2006; 1096:196-203. [PMID: 16725118 DOI: 10.1016/j.brainres.2006.04.062] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Revised: 04/04/2006] [Accepted: 04/10/2006] [Indexed: 12/29/2022]
Abstract
Existing experimental evidence suggests that PPARgamma may play a beneficial role in neuroprotection from various brain pathologies. Here we found that focal cerebral ischemia induced by middle cerebral/common carotid arteries occlusion (MCA/CCAo) induced up-regulation of PPARgamma messenger RNA in the ischemic hemisphere as early as 6 h after the ischemic event. The increased PPARgamma mRNA expression was primarily associated with neurons in the ischemic penumbra, suggesting an important role for PPARgamma in neurons after ischemia. Intraventricular injection of 15d-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), a proposed endogenous PPARgamma agonist, into the ischemic rat brains significantly increased the PPARgamma-DNA-binding activity and reduced infarction volume at 24 h after reperfusion. We propose that PPARgamma up-regulation in response to ischemia may contribute to PPARgamma activation in the presence of PPARgamma agonists. Activation of PPARgamma in neurons at an early stage after ischemia may represent a pro-survival mechanism against ischemic injury.
Collapse
Affiliation(s)
- Zhishuo Ou
- University of Texas Health Science Center-Houston, Medical School, Department of Neurology, Stroke Program, Houston, 77030, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Thulborn KR, Davis D, Snyder J, Yonas H, Kassam A. Sodium MR Imaging of Acute and Subacute Stroke for Assessment of Tissue Viability. Neuroimaging Clin N Am 2005; 15:639-53, xi-xii. [PMID: 16360594 DOI: 10.1016/j.nic.2005.08.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sodium MR imaging at 3.0 T provides high-quality images in acceptable acquisition times that allow assessment of tissue viability as defined by maintenance of sodium ion homeostasis. This application is made feasible for clinical stroke evaluation by an efficient projection pulse sequence with extremely short echo time values. This twisted projection imaging provides high signal-to-noise images at adequate resolution (5 x 5 x 5 mm(3)) in less than 10 minutes at 3.0 T. The images are quantified as tissue sodium concentration (TSC) maps that can be interpreted directly in terms of tissue viability. With infarction, baseline TSC values of less than 45 mmol/L increase at variable rates to approximately 70 mmol/L, allowing monitoring of the progression of stroke pathophysiology.
Collapse
Affiliation(s)
- Keith R Thulborn
- Center for Magnetic Resonance Research, University of Illinois at Chicago Medical Center, IL 60612, USA.
| | | | | | | | | |
Collapse
|
28
|
Storini C, Rossi E, Marrella V, Distaso M, Veerhuis R, Vergani C, Bergamaschini L, De Simoni MG. C1-inhibitor protects against brain ischemia-reperfusion injury via inhibition of cell recruitment and inflammation. Neurobiol Dis 2005; 19:10-7. [PMID: 15837556 DOI: 10.1016/j.nbd.2004.11.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 10/29/2004] [Accepted: 11/02/2004] [Indexed: 11/17/2022] Open
Abstract
Previous studies demonstrated that C1-inhibitor (C1-INH), a complement and contact-kinin systems inhibitor, is neuroprotective in cerebral ischemia. To investigate the mechanism of this action, we evaluated the expression of neurodegeneration and inflammation-related factors in mice subjected to 2-h ischemia and 2 or 46 h reperfusion. C1-INH significantly dampened the mRNA expression of the adhesion molecules P-selectin and ICAM-1 induced by the ischemic insult. It significantly decreased the pro-inflammatory cytokine (TNF alpha, IL-18) and increased the protective cytokine (IL-6, IL-10) gene expression. C1-INH treatment prevented the decrease of NFH gene, a marker of cellular integrity and counteracted the increase of pro-caspase 3, an apoptosis index. Furthermore, C1-INH markedly inhibited the activation and/or recruitment of microglia/macrophage, as shown by immunohistochemistry. In conclusion, C1-INH exerts an anti-inflammatory and anti-apoptotic action on ischemia-reperfusion injury. Our present and past data support a major effect of C1-INH on cell recruitment from the vasculature to the ischemic site.
Collapse
Affiliation(s)
- Claudio Storini
- Laboratory of Inflammation and Nervous System Diseases, Mario Negri Institute, via Eritrea, 62, 20157 Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhao X, Liu SJ, Zhang J, Strong R, Aronowski J, Grotta JC. Combining Insulin-Like Growth Factor Derivatives Plus Caffeinol Produces Robust Neuroprotection After Stroke in Rats. Stroke 2005; 36:129-34. [PMID: 15569874 DOI: 10.1161/01.str.0000149624.87661.18] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
Insulin-like growth factor-1 (IGF-1) and caffeinol are both neuroprotective and probably have different mechanisms of action; therefore, they may be more effective in combination.
Methods—
We tested the N-terminal tripeptide of IGF-1, Gly-Pro-Glu (GPE), and its analogue, G2MePE, alone and with caffeinol in a rat middle cerebral artery (MCA) suture occlusion model. We randomly assigned rats to 6 groups of 8 to 12 animals: (1) control; (2) GPE, 3 mg/kg per hour; (3) G2MePE, 0.3 mg/kg per hour; (4) caffeinol, a mixture of caffeine (10 mg/kg) with ethanol (0.32 g/kg); (5) GPE with caffeinol (combination of group 2 with 4); and (6) G2MePE with caffeinol (combination of group 3 with 4). Drugs were started 75 minutes after suture occlusion, at the start of reperfusion. Three days after MCA occlusion, neurological deficit (Neurological Deficit Score [NDS]) and lesion volume were measured.
Results—
GPE and caffeinol improved NDS by 34% and 36%, respectively (
P
<0.01), and also decreased cortical but not striatal lesion volume compared with control (GPE cortex, 121 mm
3
; caffeinol cortex, 134 mm
3
; and control, 221 mm
3
;
P
<0.01). GPE plus caffeinol did not have more efficacy than either GPE or caffeinol alone. G2MePE slightly improved NDS (19.7%,
P
=0.05) but not lesion volume. However, G2MePE plus caffeinol very significantly improved NDS (64%) and lesion volume in both cortex (combination 95 mm
3
versus control 221 mm
3
) and striatum (combination 74 mm
3
versus control 110 mm
3
) (
P
<0.001), and was significantly more effective than either caffeinol or G2MePE alone.
Conclusion—
Both GPE and caffeinol significantly protect cortex after MCA occlusion. At the doses used in this study, the GPE analogue G2MePE by itself had minimal protective effects, but when combined with caffeinol, it demonstrated robust beneficial effects on cortical and subcortical lesion size and behavioral deficit. Further study of this combination appears justified.
Collapse
Affiliation(s)
- Xiurong Zhao
- Vascular Neurology Program, Department of Neurology, University of Texas-Houston Medical School, Houston, Tex 77030, USA
| | | | | | | | | | | |
Collapse
|
30
|
Schöller K, Zausinger S, Baethmann A, Schmid-Elsaesser R. Neuroprotection in ischemic stroke—combination drug therapy and mild hypothermia in a rat model of permanent focal cerebral ischemia. Brain Res 2004; 1023:272-8. [PMID: 15374753 DOI: 10.1016/j.brainres.2004.01.094] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2004] [Indexed: 11/24/2022]
Abstract
We have recently demonstrated marked neuroprotective efficacy of a combination therapy with magnesium (calcium- and glutamate-antagonist), tirilazad (antioxidant) and mild hypothermia (MTH) in a rat model of transient focal cerebral ischemia. In the present study, we investigated MTH under conditions of permanent focal cerebral ischemia. In part I, 20 Sprague-Dawley rats were subjected to 6 h of permanent, laser-Doppler flowmetry (LDF) controlled middle cerebral artery occlusion (MCAO). Drugs were administered 30 min before and 1 h after MCAO. Hypothermia (33 degrees C) was maintained for 2 h. Infarct size was planimetrically determined after 6 h. In part II, 29 rats were assigned to the same treatment arms and subjected to 7 days of permanent MCAO. Neurological deficits and body weight were assessed daily. Infarct size was determined on day 7. In part I, MTH significantly reduced infarct formation by 52% after 6 h. In part II, high mortality within the first 3 days was observed in both groups. Treated animals showed a significantly better postoperative weight gain on day 7 and neurological recovery on days 6 and 7 compared to controls without significant differences in infarct volume. MTH seems to exert its neuroprotective properties even in a setting of permanent cerebral ischemia. High mortality and absence of infarct reduction after 7 days might be due to model limitations. Neurological recovery, the most important clinical outcome parameter, is significantly improved in 7-day survivors. Significant neuroprotection under conditions of permanent ischemia and former promising results in transient ischemia justify further investigations of MTH.
Collapse
Affiliation(s)
- Karsten Schöller
- Department of Neurosurgery, Ludwig-Maximilians-Universität, Klinikum Grosshadern, Marchioninistr. 15, 81377 Munich, Germany.
| | | | | | | |
Collapse
|
31
|
Le Belle JE, Caldwell MA, Svendsen CN. Improving the survival of human CNS precursor-derived neurons after transplantation. J Neurosci Res 2004; 76:174-83. [PMID: 15048915 DOI: 10.1002/jnr.20035] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have examined the effects of predifferentiation and energy substrate deprivation on long-term expanded human neural precursor cells (HNPCs). The pre-differentiation of HNPC cultures produced large numbers of neurons (>60%) and mature glial cells capable of generating glycogen stores that protected the neuronal population from experimental metabolic stress. When predifferentiated HNPCs were transplanted into intact adult rat hippocampus, fewer cells survived compared to undifferentiated HNPC transplants. This cell death was completely attenuated, however, when predifferentiated HNPC cultures were pretreated to boost glial energy stores and resulted in greatly increased neuronal survival in vivo. The transplanted cells primarily engrafted within the granular layer of the dentate gyrus, where a large proportion of the predifferentiated HNPCs co-expressed neuronal markers whereas most HNPCs outside of the neuronal layer did not, indicating that the predifferentiated cells remained capable of responding to local cues in the adult brain. Undifferentiated HNPCs migrated more widely in the brain after grafting than did the predifferentiated cells, which generally remained within the hippocampus.
Collapse
Affiliation(s)
- J E Le Belle
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom.
| | | | | |
Collapse
|
32
|
Lei B, Popp S, Capuano-Waters C, Cottrell JE, Kass IS. Lidocaine attenuates apoptosis in the ischemic penumbra and reduces infarct size after transient focal cerebral ischemia in rats. Neuroscience 2004; 125:691-701. [PMID: 15099683 DOI: 10.1016/j.neuroscience.2004.02.034] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2004] [Indexed: 11/29/2022]
Abstract
Lidocaine is a local anesthetic and antiarrhythmic agent. Although clinical and experimental studies have shown that an antiarrhythmic dose of lidocaine can protect the brain from ischemic damage, the underlying mechanisms are unknown. In the present study, we examined whether lidocaine inhibits neuronal apoptosis in the penumbra in a rat model of transient focal cerebral ischemia. Male Wistar rats underwent a 90-min temporary occlusion of middle cerebral artery. Lidocaine was given as an i.v. bolus (1.5 mg/kg) followed by an i.v. infusion (2 mg/kg/h) for 180 min, starting 30 min before ischemia. Rats were killed and brain samples were collected at 4 and 24 h after ischemia. Apoptotic changes were evaluated by immunohistochemistry for cytochrome c release and caspase-3 activation and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) for DNA fragmentation. Cytochrome c release and caspase-3 activation were detected at 4 and 24 h after ischemia and DNA fragmentation was detected at 24 h. Double-labeling with NeuN, a neuronal marker, demonstrated that cytochrome c, caspase-3, and TUNEL were confined to neurons. Lidocaine reduced cytochrome c release and caspase-3 activation in the penumbra at 4 h and diminished DNA fragmentation in the penumbra at 24 h. Lidocaine treatment improved early electrophysiological recovery and reduced the size of the cortical infarct at 24 h, but had no significant effect on cerebral blood flow in either the penumbra or core during ischemia. These findings suggest that lidocaine attenuates apoptosis in the penumbra after transient focal cerebral ischemia. The infarct-reducing effects of lidocaine may be due, in part, to the inhibition of apoptotic cell death in the penumbra.
Collapse
Affiliation(s)
- B Lei
- Department of Anesthesiology, State University of New York Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | | | | | | | | |
Collapse
|
33
|
Zausinger S, Schöller K, Plesnila N, Schmid-Elsaesser R. Combination drug therapy and mild hypothermia after transient focal cerebral ischemia in rats. Stroke 2003; 34:2246-51. [PMID: 12893947 DOI: 10.1161/01.str.0000083622.65684.21] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE We have recently demonstrated that pretreatment with magnesium (calcium and glutamate antagonist) and tirilazad (antioxidant) in combination with intraischemic mild hypothermia (33 degrees C) (MTH) offers superior neuroprotective efficacy in a rat model of focal transient cerebral ischemia. In the present study, we investigated the time window of this treatment strategy with a posttreatment regimen to define its role for stroke patients. METHODS We subjected 48 Sprague-Dawley rats to 90 minutes of middle cerebral artery occlusion by an intraluminal filament. Bilateral regional cerebral blood flow was continuously recorded by laser Doppler flowmetry. Combination therapy with MTH was started at 0, 1, 3, and 5 hours after induction of ischemia. Drugs were given in 1-hour intervals, and hypothermia was maintained for 2 hours. Neurological deficits were assessed daily. Infarct size was planimetrically determined on postoperative day 7. RESULTS Combination therapy with MTH significantly reduced infarct volume compared with normothermic controls by -74%, -49%, and -45% when applied at 0, 1, and 3 hours after induction of ischemia. Furthermore, these treatment groups showed less neurological deficits on postischemic days 1 and 2 (P<0.05). Onset of treatment 5 hours after middle cerebral artery occlusion failed to significantly reduce infarct formation and neurological deficits. CONCLUSIONS The therapeutic window of the new combination therapy is at least 3 hours after onset of ischemia, comparable to that of moderate hypothermia (30 degrees C), a grade of hypothermia associated with higher risks of severe side effects.
Collapse
Affiliation(s)
- Stefan Zausinger
- Department of Neurosurgery, Ludwig-Maximilians-Universität, Klinikum Grosshadern, Munich, Germany.
| | | | | | | |
Collapse
|
34
|
Aronowski J, Labiche LA. Perspectives on reperfusion-induced damage in rodent models of experimental focal ischemia and role of gamma-protein kinase C. ILAR J 2003; 44:105-9. [PMID: 12652005 DOI: 10.1093/ilar.44.2.105] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Ischemic stroke represents the leading cause of death and disability among elderly people. Most stroke survivors are left with lifelong disability. With the exception of tissue-type plasminogen activator (t-PA), no effective therapy exists for the management of acute stroke. Understanding the role of various extrinsic and intrinsic pathogenic factors of ischemic damage represents a prime objective of ongoing stroke research. An important variable affecting stroke outcome is the presence or absence of reperfusion (recanalization of the occluded vessel) following an ischemic event. It appears that early reperfusion after a stroke is beneficial and capable of reversing the majority of ischemic dysfunctions. However, in some instances, late reperfusion may contrarily trigger deleterious processes and lead to more ischemic damage. Examples of ischemia/reperfusion damage using an experimental model of focal ischemia in rodents are provided, along with evidence that the brain-enriched gamma-isoform of protein kinase C may represent an important mediator of reperfusion-induced brain injury in mutant mice.
Collapse
Affiliation(s)
- Jaroslaw Aronowski
- Department of Neurology, University of Texas-Houston Medical School, Houston, TX, USA
| | | |
Collapse
|
35
|
Aronowski J, Strong R, Shirzadi A, Grotta JC. Ethanol plus caffeine (caffeinol) for treatment of ischemic stroke: preclinical experience. Stroke 2003; 34:1246-51. [PMID: 12690223 DOI: 10.1161/01.str.0000068170.80517.b3] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Ethanol and caffeine are 2 common psychoactive dietary components. We have recently shown that low-dose ethanol plus caffeine results in a 70% to 80% reduction of infarct volume after reversible common carotid/middle cerebral artery (CCA/MCA) occlusion in rats. The combination (caffeinol) was effective after either oral pretreatment or intravenous administration starting up to 2 hours after stroke onset. Ethanol alone aggravated ischemic damage, while caffeine alone was without effect. Daily caffeinol for 2 weeks before ischemia eliminated the neuroprotection seen with acute treatment (tolerance). The purpose of our present study was to further characterize the properties of caffeinol as a possible treatment for ischemic stroke. METHODS The transient CCA/MCA occlusion model was used in all experiments. Five sets of experiments were conducted (1) to test the effectiveness of various doses of ethanol (0.2 to 0.65 g/kg) and caffeine (3 to 10 mg/kg) in the caffeinol mixture; (2) to test whether the neuroprotective dose of caffeinol can improve behavioral dysfunction; (3) to test whether chronic ethanol or caffeine before ischemia will affect efficacy of caffeinol treatment; (4) to test whether the protective effect of caffeinol can be improved by pairing it with 35 degrees C hypothermia; and (5) to test whether caffeinol affects frequency of hemorrhage after administration of recombinant tissue plasminogen activator (rtPA) in ischemic animals. RESULTS Doses as low as 0.2 g/kg of ethanol and 6 mg/kg of caffeine in the caffeinol were effective in reducing cortical infarct volume and behavioral dysfunction after transient CCA/MCA occlusion. Daily exposure to ethanol but not caffeine before CCA/MCA occlusion eliminated the therapeutic efficacy of acute caffeinol treatment, similar to the tolerance observed after chronic exposure to caffeinol. The therapeutic effect of caffeinol could be further improved by pairing it with mild intraischemic hypothermia, and caffeinol did not increase hemorrhagic infarction when given in combination with rtPA. CONCLUSIONS Low doses of caffeinol, equivalent to no more than 2 to 3 cups of strong coffee and 1 cocktail, are consistently and highly neuroprotective, are well tolerated, can be added to other therapies to increase the effect of each, and do not interfere with or complicate rtPA therapy. Caffeinol is an appropriate candidate for clinical trial in stroke patients, although it may be less effective in patients with regular alcohol intake.
Collapse
Affiliation(s)
- Jaroslaw Aronowski
- Stroke Program, Department of Neurology, University of Texas at Houston Medical School, Houston, 77030, USA.
| | | | | | | |
Collapse
|
36
|
Sanchez V, Zeini M, Camarero J, O'Shea E, Bosca L, Green AR, Colado MI. The nNOS inhibitor, AR-R17477AR, prevents the loss of NF68 immunoreactivity induced by methamphetamine in the mouse striatum. J Neurochem 2003; 85:515-24. [PMID: 12675928 DOI: 10.1046/j.1471-4159.2003.01714.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The present study examined the time-course and regionally-selective changes in the levels of the neurofilament protein NF68 in the mouse brain induced by methamphetamine (METH). The ability of low ambient temperature, or of the specific neuronal nitric oxide synthase (nNOS) inhibitor AR-R17477AR, to protect against both long-term striatal NF68 and dopamine loss induced by METH (3 mg/kg, i.p.) was also studied. Seven days after METH administration (3, 6 and 9 mg/kg, i.p., three times at 3 h intervals), mice showed a reduction of about 40% in immunoreactivity for NF68 in the striatum. This effect was not produced in cortex after METH administration at the dose of 3 mg/kg. No difference from controls was observed when measurements were carried out 1 h and 24 h after the last METH injection at the dose of 3 mg/kg. The loss of NF68 immunoreactivity seems to be associated with the long-term dopamine depletion induced by METH, since no change in serotonin concentration is observed in either the striatum or cortex 7 days after dosing. Animals kept at a room temperature of 4 degrees C showed a loss of NF68 similar to those treated at 22 degrees C but an attenuation of dopamine depletion in the striatum. Pre-treatment with AR-R17477AR (5 mg/kg, s.c.) 30 min before each of the three METH (3 mg/kg, i.p.) injections provided complete protection against METH-induced loss of NF68 immunoreactivity and attenuated the decrease in striatal dopamine and HVA concentrations by about 50%. These data indicate that both the reduction of NF68 immunoreactivity and the loss of dopamine concentration are due to an oxidative stress process mediated by reactive nitrogen species, and are not due to changes in body temperature.
Collapse
Affiliation(s)
- Veronica Sanchez
- Departamento de Farmacologia and Facultad de Medicina Instituto de Bioquimica CSIC-UCM, Facultad de Farmacia, Universidad Complutense, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
37
|
Zhu DY, Deng Q, Yao HH, Wang DC, Deng Y, Liu GQ. Inducible nitric oxide synthase expression in the ischemic core and penumbra after transient focal cerebral ischemia in mice. Life Sci 2002; 71:1985-96. [PMID: 12175893 DOI: 10.1016/s0024-3205(02)01970-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present observations examined the hypothesis that the iNOS expression in the ischemic penumbra after a transient focal ischemic insult is involved in the recruitment of penumbra into infarction. The middle cerebral artery in mice was occluded for 2 h by an intraluminal filament and then recirculated. The measurement of iNOS activity, iNOS protein formation and NO concentration in the ischemic core and penumbra, and the determination of infarct volume were performed at 6, 12, 24 and 48 h after reperfusion. iNOS protein and iNOS enzymatic activity appeared at 6 h, peaked at 24 h, and declined at 48 h in the penumbra after reperfusion. iNOS protein was not detectable in contralateral area and in sham-operated brains. The time course of iNOS protein, enzymatic activity and NO concentration in the penumbra but not in the core matched the process of infarct maturation. Treatment with iNOS inhibitor aminoguanidine (100 mg.kg(-1), i.p.) at 6 and 12 h after reperfusion inhibited iNOS activity by 88.0 +/- 10.4% and reduced NO concentration by 48.5 +/- 8.3% in the penumbra, and lessened infarct size by 48.8 +/- 7.2%. The iNOS activity and NO level in the core were not affected by the administration of aminoguanidine. These results suggest that iNOS expression in the ischemic penumbra is involved in the recruitment of penumbra into infarction and thereby contributing to the enlargement of infarct.
Collapse
Affiliation(s)
- Dong-Ya Zhu
- Pharmacology Department, New Drug Research Center, China Pharmaceutical University, Tong Jia Xiang 24#, Nanjing 210009, China.
| | | | | | | | | | | |
Collapse
|
38
|
Leker RR, Shohami E. Cerebral ischemia and trauma-different etiologies yet similar mechanisms: neuroprotective opportunities. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2002; 39:55-73. [PMID: 12086708 DOI: 10.1016/s0165-0173(02)00157-1] [Citation(s) in RCA: 285] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cerebral ischemia leads to brain damage caused by pathogenetic mechanisms that are also activated by neurotrauma. These mechanisms include among others excitotoxicity, over production of free radicals, inflammation and apoptosis. Furthermore, cerebral ischemia and trauma both trigger similar auto-protective mechanisms including the production of heat shock proteins, anti-inflammatory cytokines and endogenous antioxidants. Neuroprotective therapy aims at minimizing the activation of toxic pathways and at enhancing the activity of endogenous neuroprotective mechanisms. The similarities in the damage-producing and endogenous auto-protective mechanisms may imply that neuroprotective compounds found to be active against one of these conditions may indeed be also protective in the other. This review summarizes the pathogenetic events of ischemic and traumatic brain injury and reviews the neuroprotective strategies employed thus far in each of these conditions with a special emphasize on their clinical relevance and on future directions in the field of neuronal protection.
Collapse
Affiliation(s)
- Ronen R Leker
- Department of Neurology and the Agnes Ginges Center for Human Neurogenetics, Hebrew University-Hadassah Medical School and Hadassah University Hospital, Jerusalem, Israel.
| | | |
Collapse
|
39
|
Zausinger S, Baethmann A, Schmid-Elsaesser R. Anesthetic methods in rats determine outcome after experimental focal cerebral ischemia: mechanical ventilation is required to obtain controlled experimental conditions. BRAIN RESEARCH. BRAIN RESEARCH PROTOCOLS 2002; 9:112-21. [PMID: 12034330 DOI: 10.1016/s1385-299x(02)00138-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Anesthetic agents, pH, blood gases and blood pressure have all been found to influence the pathophysiology of experimental stroke. In experimental research, rats are predominantly used to investigate the effects of focal cerebral ischemia. Chloral hydrate, applied intraperitoneally (i.p.), and halothane, applied via face-mask in spontaneously breathing animals or via endotracheal tube in mechanically ventilated animals are popular methods of anesthesia. We investigated the potential of these anesthetic methods to maintain physiologic conditions during focal cerebral ischemia and their influence on postischemic mortality and histological outcome. METHODS Thirty male Sprague-Dawley rats were subjected to 90 min of middle cerebral artery occlusion by insertion of an intraluminal thread and assigned to one of three groups (n=10 each): (A) chloral hydrate i.p./spontaneously breathing; (B) halothane in 70:30 (%) N2O/O2 via face-mask/spontaneously breathing; and (C) halothane in 70:30 (%) N2O/O2 via endotracheal tube/mechanically ventilated. Physiologic parameters were measured before, during, and after ischemia. Infarct volume was histologically assessed after 7 days. RESULTS All anesthetic techniques except mechanical ventilation via an endotracheal tube resulted in considerably fluctuating blood gases levels, hypercapnia, acidosis and low blood pressure. All spontaneously breathing animals (groups A and B) exhibited a higher postischemic mortality and significantly larger infarct volumes than group C with intubated and ventilated animals. CONCLUSIONS Intra- and postischemic physiologic parameters such as blood pressure, pH, and blood gases critically determine outcome after focal cerebral ischemia. Although anesthesia by halothane via face-mask allowed better control of depth of anesthesia than chloral hydrate, we have found this method to be unsatisfactory due to insufficient control of ventilation and waste of anesthetic gases. Experiments with rats requiring normal physiologic parameters should be performed under conditions of controlled mechanical ventilation and sufficient analgesia.
Collapse
Affiliation(s)
- S Zausinger
- Department of Neurosurgery, Ludwig-Maximilians-Universität, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany.
| | | | | |
Collapse
|
40
|
Lauer KK, Shen H, Stein EA, Ho KC, Kampine JP, Hudetz AG. Focal cerebral ischemia in rats produced by intracarotid embolization with viscous silicone. Neurol Res 2002; 24:181-90. [PMID: 11877903 DOI: 10.1179/016164102101199594] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Many factors contribute to the severity of neuronal cell death and the functional outcome in stroke. We describe an embolic model of focal cerebral ischemia in the rat that does not require craniotomy and is compatible with continuous measurement of regional CBF using multichannel laser Doppler flow (LDF) technique. Either a 22 microliters (large lesion) or 11 microliters (small lesion) bolus of viscous silicone was injected cephalad into the internal carotid artery. Upon injection, LDF decreased abruptly, most severely in the parietal cortex (-74% +/- 5%) in the large lesion and in the occipital cortex (-69% +/- 10%) in the small lesion model. Over the first hour, post-embolization LDF improved in most areas (e.g. -48% +/- 9% parietal, large lesion) but declined in the small lesion group in the occipital region (-81% +/- 8%). CBF measured by [C]14-IAP autoradiography 1 h post-embolization in the large lesion model demonstrated near-hemispheric ischemia (70% of hemisphere) with sparing of cingulate cortex. Autoradiography demonstrated that ischemia in the small lesion was largely cortical. Light microscopy of brains embolized with 11 microliters of dyed silicone showed filling of pial vessels with no silicone in the Circle of Willis or parenchyma. No animals in the large lesion group survived 24 h. Thirteen of 15 animals in the small lesion group survived for two weeks with resolution of initial hemiplegia, ocular asymmetry and weight loss. Hematoxylin-eosin staining two weeks post-embolization showed signs of severe hypoxia and infarction. In conclusion, the intracarotid silicone embolization technique produces a titrable, reproducible permanent ischemic injury by blocking perfusion in the pial circulation, and is amenable to multisite monitoring with laser Doppler flowmetry. The smaller embolus produces cortical infarction with high rate of survival and neurological recovery.
Collapse
Affiliation(s)
- Kathryn K Lauer
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Kupina NC, Nath R, Bernath EE, Inoue J, Mitsuyoshi A, Yuen PW, Wang KK, Hall ED. The novel calpain inhibitor SJA6017 improves functional outcome after delayed administration in a mouse model of diffuse brain injury. J Neurotrauma 2001; 18:1229-40. [PMID: 11721741 DOI: 10.1089/089771501317095269] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A principal mechanism of calcium-mediated neuronal injury is the activation of neutral proteases known as calpains. Proteolytic substrates for calpain include receptor and cytoskeletal proteins, signal transduction enzymes and transcription factors. Recently, calpain inhibitors have been shown to provide benefit in rat models of focal head injury and focal cerebral ischemia. The present study sought to investigate, in experiment 1, the time course of calpain-mediated cytoskeletal injury in a mouse model of diffuse head injury by measuring the 150- and 145-kDa alpha-spectrin breakdown products (SBDP). Secondly, in experiment 2, we examined the effect of early (20 min postinjury) administration of the novel calpain inhibitor SJA6017 on functional outcome measured 24 h following injury and its effect on posttraumatic alpha-spectrin degradation. Lastly, in experiment 3, we examined the effect of delayed (4 or 6 h postinjury) administration of SJA6017 on 24-h postinjury functional outcome. In experiment 1, isoflurane-anesthetized male CF-1 mice (18-22 g) were subjected to a 750 g-cm weight drop-induced injury and were sacrificed for SBDP analysis at postinjury times of 30 min, and 1, 2, 6, 24 and 48 h (plus sham). In experiments 2 and 3, mice were injured as described, and delivered a single tail vein injection of either SJA6017 (0.3, 1, or 3 mg/kg) or vehicle (administered immediately, 4 or 6 h postinjury [3 mg/kg]). Functional outcome was evaluated in both studies, and, in experiment 2, 24-h postinjury assessment of SBDPs was determined. Following injury, the level of SBDP 145 was significantly different from sham at 24 and 48 h in cortical and at 24 h in the hippocampal tissues and at 48 h in the striatum. Immediate postinjury administration of SJA6017 resulted in a dose-related improvement in 24-h functional outcome (p < 0.05 at 3 mg/kg). Significance was maintained after a 4-h delay of the 3 mg/kg, but was lost after a 6-h delay. Despite improvement in functional outcome at 24 h, SJA6017 did not reduce spectrin breakdown in cortical or hippocampal tissues. These results support a role for calpain-mediated neuronal injury and the potential for a practical therapeutic window for calpain inhibition following traumatic brain injury. However, measurements of regional spectrin degradation may not be the most sensitive marker for determining the effects of calpain inhibition.
Collapse
Affiliation(s)
- N C Kupina
- Neuroscience Therapeutics, Pfizer Global Research and Development-Ann Arbor Laboratories, Michigan 48015, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Reglodi D, Somogyvari-Vigh A, Vigh S, Kozicz T, Arimura A. Delayed systemic administration of PACAP38 is neuroprotective in transient middle cerebral artery occlusion in the rat. Stroke 2000; 31:1411-7. [PMID: 10835464 DOI: 10.1161/01.str.31.6.1411] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Many substances have been shown to reduce brain damage in models of stroke, but mainly when given either before or shortly after the onset of ischemia. Delayed systemic administration of pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to attenuate the neuronal damage in the hippocampus in a model of global ischemia in rats. The present study examined the neuroprotective action of delayed systemic administration of PACAP38 in a model of transient focal ischemia produced by middle cerebral artery occlusion (MCAO) in rats. METHODS We administered PACAP38 as an intravenous bolus (20 nmol/kg body wt) followed by an intravenous infusion for 48 hours using a micro-osmotic pump at a rate of 160 pmol/microL per hour, beginning 4, 8, or 12 hours after a 2-hour transient MCAO using a filament model. The size of the infarct was determined by examining 2-mm-thick brain sections stained with triphenyltetrazolium chloride, followed by image analysis. Control animals received intravenously 0.1% bovine serum albumin in 0.9% saline as a bolus and infusion at the same time intervals. RESULTS The administration of PACAP38 beginning 4 hours after MCAO significantly reduced the infarct size by 50.88%. Treatment with PACAP38 starting 8 or 12 hours after the onset of ischemia did not result in a significant reduction of the infarct size, although infarct volumes tended to be smaller than in the control groups. CONCLUSIONS Systemic administration of PACAP38 should be clinically useful for reducing brain damage resulting from stroke even when administration is delayed for several hours.
Collapse
MESH Headings
- Animals
- Brain Damage, Chronic/etiology
- Brain Damage, Chronic/pathology
- Brain Damage, Chronic/prevention & control
- Drug Administration Schedule
- Drug Evaluation, Preclinical
- Infarction, Middle Cerebral Artery/complications
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/pathology
- Injections, Intravenous
- Ischemic Attack, Transient/drug therapy
- Ischemic Attack, Transient/etiology
- Ischemic Attack, Transient/pathology
- Male
- Neuropeptides/administration & dosage
- Neuropeptides/therapeutic use
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/therapeutic use
- Pituitary Adenylate Cyclase-Activating Polypeptide
- Rats
- Time Factors
Collapse
Affiliation(s)
- D Reglodi
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70037, USA
| | | | | | | | | |
Collapse
|