51
|
Chen R, Lai UH, Zhu L, Singh A, Ahmed M, Forsyth NR. Reactive Oxygen Species Formation in the Brain at Different Oxygen Levels: The Role of Hypoxia Inducible Factors. Front Cell Dev Biol 2018; 6:132. [PMID: 30364203 PMCID: PMC6192379 DOI: 10.3389/fcell.2018.00132] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/21/2018] [Indexed: 12/11/2022] Open
Abstract
Hypoxia inducible factor (HIF) is the master oxygen sensor within cells and is central to the regulation of cell responses to varying oxygen levels. HIF activation during hypoxia ensures optimum ATP production and cell integrity, and is associated both directly and indirectly with reactive oxygen species (ROS) formation. HIF activation can either reduce ROS formation by suppressing the function of mitochondrial tricarboxylic acid cycle (TCA cycle), or increase ROS formation via NADPH oxidase (NOX), a target gene of HIF pathway. ROS is an unavoidable consequence of aerobic metabolism. In normal conditions (i.e., physioxia), ROS is produced at minimal levels and acts as a signaling molecule subject to the dedicated balance between ROS production and scavenging. Changes in oxygen concentrations affect ROS formation. When ROS levels exceed defense mechanisms, ROS causes oxidative stress. Increased ROS levels can also be a contributing factor to HIF stabilization during hypoxia and reoxygenation. In this review, we systemically review HIF activation and ROS formation in the brain during hypoxia and hypoxia/reoxygenation. We will then explore the literature describing how changes in HIF levels might provide pharmacological targets for effective ischaemic stroke treatment. HIF accumulation in the brain via HIF prolyl hydroxylase (PHD) inhibition is proposed as an effective therapy for ischaemia stroke due to its antioxidation and anti-inflammatory properties in addition to HIF pro-survival signaling. PHD is a key regulator of HIF levels in cells. Pharmacological inhibition of PHD increases HIF levels in normoxia (i.e., at 20.9% O2 level). Preconditioning with HIF PHD inhibitors show a neuroprotective effect in both in vitro and in vivo ischaemia stroke models, but post-stroke treatment with PHD inhibitors remains debatable. HIF PHD inhibition during reperfusion can reduce ROS formation and activate a number of cellular survival pathways. Given agents targeting individual molecules in the ischaemic cascade (e.g., antioxidants) fail to be translated in the clinic setting, thus far, HIF pathway targeting and thereby impacting entire physiological networks is a promising drug target for reducing the adverse effects of ischaemic stroke.
Collapse
Affiliation(s)
- Ruoli Chen
- School of Pharmacy, Keele University, Staffordshire, United Kingdom.,Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - U Hin Lai
- School of Pharmacy, Keele University, Staffordshire, United Kingdom
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Ayesha Singh
- School of Pharmacy, Keele University, Staffordshire, United Kingdom.,Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Muhammad Ahmed
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom.,College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Nicholas R Forsyth
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
52
|
Gaidhani N, Uteshev VV. Treatment duration affects cytoprotective efficacy of positive allosteric modulation of α7 nAChRs after focal ischemia in rats. Pharmacol Res 2018; 136:121-132. [PMID: 30205140 PMCID: PMC6218269 DOI: 10.1016/j.phrs.2018.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/22/2018] [Accepted: 09/02/2018] [Indexed: 12/30/2022]
Abstract
To minimize irreversible brain injury after acute ischemic stroke (AIS), the time to treatment (i.e., treatment delay) should be minimized. However, thus far, all cytoprotective clinical trials have failed. Analysis of literature identified short treatment durations (≤72 h) as a common motif among completed cytoprotective clinical trials. Here, we argue that short cytoprotective regimens even if given early after AIS may only slow down the evolution of ischemic brain injury and fail to deliver sustained long-term solutions leading to relapses that may be misinterpreted for conceptual failure of cytoprotection. In this randomized blinded study, we used young adult male rats subjected to transient 90 min suture middle cerebral artery occlusion (MCAO) and treated with acute vs. sub-chronic regimens of PNU120596, a prototypical positive allosteric modulator of α7 nicotinic acetylcholine receptors with anti-inflammatory cytoprotective properties to test the hypothesis that insufficient treatment durations may reduce therapeutic benefits of otherwise efficacious cytoprotectants after AIS. A single acute treatment 90 min after MCAO significantly reduced brain injury and neurological deficits 24 h later, but these effects vanished 72 h after MCAO. These relapses were avoided by utilizing sub-chronic treatments. Thus, extending treatment duration augments therapeutic efficacy of PNU120596 after MCAO. Furthermore, sub-chronic treatments could offset the negative effects of prolonged treatment delays in cases where the acute treatment window after MCAO was left unexploited. We conclude that a combination of short treatment delays and prolonged treatment durations may be required to maximize therapeutic effects of PNU120596, reduce relapses and ensure sustained therapeutic efficacy after AIS. Similar concepts may hold for other cytoprotectants including those that failed in clinical trials.
Collapse
Affiliation(s)
- Nikhil Gaidhani
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States.
| |
Collapse
|
53
|
Jiménez-Xarrié E, Pérez B, Dantas AP, Puertas-Umbert L, Martí-Fabregas J, Chamorro Á, Planas AM, Vila E, Jiménez-Altayó F. Uric Acid Treatment After Stroke Prevents Long-Term Middle Cerebral Artery Remodelling and Attenuates Brain Damage in Spontaneously Hypertensive Rats. Transl Stroke Res 2018; 11:1332-1347. [PMID: 30219993 DOI: 10.1007/s12975-018-0661-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/02/2018] [Accepted: 08/30/2018] [Indexed: 01/21/2023]
Abstract
Hypertension is the most important modifiable risk factor for stroke and is associated with poorer post-stroke outcomes. The antioxidant uric acid is protective in experimental normotensive ischaemic stroke. However, it is unknown whether this treatment exerts long-term protection in hypertension. We aimed to evaluate the impact of transient intraluminal middle cerebral artery (MCA) occlusion (90 min)/reperfusion (1-15 days) on brain and vascular damage progression in adult male Wistar-Kyoto (WKY; n = 36) and spontaneously hypertensive (SHR; n = 37) rats treated (i.v./120 min post-occlusion) with uric acid (16 mg kg-1) or vehicle (Locke's buffer). Ischaemic brain damage was assessed longitudinally with magnetic resonance imaging and properties of MCA from both hemispheres were studied 15 days after stroke. Brain lesions in WKY rats were associated with a transitory increase in circulating IL-18 and cerebrovascular oxidative stress that did not culminate in long-term MCA alterations. In SHR rats, more severe brain damage and poorer neurofunctional outcomes were coupled to higher cortical cerebral blood flow at the onset of reperfusion, a transient increase in oxidative stress and long-lasting stroke-induced MCA hypertrophic remodelling. Thus, stroke promotes larger brain and vascular damage in hypertensive rats that persists for long-time. Uric acid administered during early reperfusion attenuated short- and long-term brain injuries in both normotensive and hypertensive rats, an effect that was associated with abolishment of the acute oxidative stress response and prevention of stroke-induced long-lasting MCA remodelling in hypertension. These results suggest that uric acid might be an effective strategy to improve stroke outcomes in hypertensive subjects.
Collapse
Affiliation(s)
- Elena Jiménez-Xarrié
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Instituto de Investigación Biomédica (IIB)-Sant Pau, Barcelona, Spain
| | - Belén Pérez
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - Ana Paula Dantas
- Institut Clínic Cardiovascular, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lídia Puertas-Umbert
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - Joan Martí-Fabregas
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Instituto de Investigación Biomédica (IIB)-Sant Pau, Barcelona, Spain
| | - Ángel Chamorro
- Comprehensive Stroke Center, Hospital Clínic, University of Barcelona, Barcelona, Spain.,Àrea de Neurociènces, IDIBAPS, Barcelona, Spain
| | - Anna Maria Planas
- Àrea de Neurociènces, IDIBAPS, Barcelona, Spain.,Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Elisabet Vila
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - Francesc Jiménez-Altayó
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain.
| |
Collapse
|
54
|
Goyal M, Ganesh A, Brown S, Menon BK, Hill MD. Suggested modification of presentation of stroke trial results. Int J Stroke 2018; 13:669-672. [DOI: 10.1177/1747493018778122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The modified Rankin Scale (mRS) at 90 days after stroke onset has become the preferred outcome measure in acute stroke trials, including recent trials of interventional therapies. Reporting the range of modified Rankin Scale scores as a paired horizontal stacked bar graph (colloquially known as “Grotta bars”) has become the conventional method of visualizing modified Rankin Scale results. Grotta bars readily illustrate the levels of the ordinal modified Rankin Scale in which benefit may have occurred. However, complementing the available graphical information by including additional features to convey statistical significance may be advantageous. We propose a modification of the horizontal stacked bar graph with illustrative examples. In this suggested modification, the line joining the segments of the bar graph (e.g. modified Rankin Scale 1–2 in treatment arm to modified Rankin Scale 1–2 in control arm) is given a color and thickness based on the p-value of the result at that level (in this example, the p-value of modified Rankin Scale 0–1 vs. 2–6)—a thick green line for p-values <0.01, thin green for p-values of 0.01 to <0.05, gray for 0.05 to <0.10, thin red for 0.10 to <0.90, and thick red for p-values ≥0.90 or outcome favoring the control group. Illustrative examples from four recent trials (ESCAPE, SWIFT-PRIME, IST-3, ASTER) are shown to demonstrate the range of significant and non-significant effects that can be captured using this proposed method. By formalizing a display of outcomes which includes statistical tests of all possible dichotomizations of the Rankin scale, this approach also encourages pre-specification of such hypotheses. Prespecifying tests of all six dichotomizations of the Rankin scale provides all possible statistical information in an a priori fashion. Since the result of our proposed approach is six distinct dichotomized tests in addition to a primary test, e.g. of the ordinal Rankin shift, it may be prudent to account for multiplicity in testing by using dichotomized p-values only after adjustment, such as by the Bonferroni or Hochberg-Holm methods. Whether p-values are nominal or adjusted may be left to the discretion of the presenter as long as the presence or absence is clearly stated in the statistical methods. Our proposed modification results in a visually intuitive summary of both the size of the effect—represented by the matched bars and their connecting segments—as well as its statistical relevance.
Collapse
Affiliation(s)
- Mayank Goyal
- Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Aravind Ganesh
- Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Scott Brown
- Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
- Altair Biostatistics, Mooresville, USA
| | - Bijoy K Menon
- Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Michael D Hill
- Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
| |
Collapse
|
55
|
Chen HS, Chen X, Li WT, Shen JG. Targeting RNS/caveolin-1/MMP signaling cascades to protect against cerebral ischemia-reperfusion injuries: potential application for drug discovery. Acta Pharmacol Sin 2018; 39:669-682. [PMID: 29595191 PMCID: PMC5943912 DOI: 10.1038/aps.2018.27] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/26/2018] [Indexed: 02/07/2023] Open
Abstract
Reactive nitrogen species (RNS) play important roles in mediating cerebral ischemia-reperfusion injury. RNS activate multiple signaling pathways and participate in different cellular events in cerebral ischemia-reperfusion injury. Recent studies have indicated that caveolin-1 and matrix metalloproteinase (MMP) are important signaling molecules in the pathological process of ischemic brain injury. During cerebral ischemia-reperfusion, the production of nitric oxide (NO) and peroxynitrite (ONOO−), two representative RNS, down-regulates the expression of caveolin-1 (Cav-1) and, in turn, further activates nitric oxide synthase (NOS) to promote RNS generation. The increased RNS further induce MMP activation and mediate disruption of the blood-brain barrier (BBB), aggravating the brain damage in cerebral ischemia-reperfusion injury. Therefore, the feedback interaction among RNS/Cav-1/MMPs provides an amplified mechanism for aggravating ischemic brain damage during cerebral ischemia-reperfusion injury. Targeting the RNS/Cav-1/MMP pathway could be a promising therapeutic strategy for protecting against cerebral ischemia-reperfusion injury. In this mini-review article, we highlight the important role of the RNS/Cav-1/MMP signaling cascades in ischemic stroke injury and review the current progress of studies seeking therapeutic compounds targeting the RNS/Cav-1/MMP signaling cascades to attenuate cerebral ischemia-reperfusion injury. Several representative natural compounds, including calycosin-7-O-β-D-glucoside, baicalin, Momordica charantia polysaccharide (MCP), chlorogenic acid, lutein and lycopene, have shown potential for targeting the RNS/Cav-1/MMP signaling pathway to protect the brain in ischemic stroke. Therefore, the RNS/Cav-1/MMP pathway is an important therapeutic target in ischemic stroke treatment.
Collapse
|
56
|
Xiong XY, Liu L, Yang QW. Refocusing Neuroprotection in Cerebral Reperfusion Era: New Challenges and Strategies. Front Neurol 2018; 9:249. [PMID: 29740385 PMCID: PMC5926527 DOI: 10.3389/fneur.2018.00249] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Pathophysiological processes of stroke have revealed that the damaged brain should be considered as an integral structure to be protected. However, promising neuroprotective drugs have failed when translated to clinical trials. In this review, we evaluated previous studies of neuroprotection and found that unsound patient selection and evaluation methods, single-target treatments, etc., without cerebral revascularization may be major reasons of failed neuroprotective strategies. Fortunately, this may be reversed by recent advances that provide increased revascularization with increased availability of endovascular procedures. However, the current improved effects of endovascular therapy are not able to match to the higher rate of revascularization, which may be ascribed to cerebral ischemia/reperfusion injury and lacking of neuroprotection. Accordingly, we suggest various research strategies to improve the lower therapeutic efficacy for ischemic stroke treatment: (1) multitarget neuroprotectant combinative therapy (cocktail therapy) should be investigated and performed based on revascularization; (2) and more efforts should be dedicated to shifting research emphasis to establish recirculation, increasing functional collateral circulation and elucidating brain–blood barrier damage mechanisms to reduce hemorrhagic transformation. Therefore, we propose that a comprehensive neuroprotective strategy before and after the endovascular treatment may speed progress toward improving neuroprotection after stroke to protect against brain injury.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
57
|
Antonic A, Dottori M, Macleod MR, Donnan GA, Howells DW. NXY-059, a Failed Stroke Neuroprotectant, Offers No Protection to Stem Cell-Derived Human Neurons. J Stroke Cerebrovasc Dis 2018; 27:2158-2165. [PMID: 29673616 DOI: 10.1016/j.jstrokecerebrovasdis.2018.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Developing new medicines is a complex process where understanding the reasons for both failure and success takes us forward. One gap in our understanding of most candidate stroke drugs before clinical trial is whether they have a protective effect on human tissues. NXY-059 is a spin-trap reagent hypothesized to have activity against the damaging oxidative biology which accompanies ischemic stroke. Re-examination of the preclinical in vivo dataset for this agent in the wake of the failed SAINT-II RCT highlighted the presence of a range of biases leading to overestimation of the magnitude of NXY-059's effects in laboratory animals. Therefore, NXY-059 seemed an ideal candidate to evaluate in human neural tissues to determine whether human tissue testing might improve screening efficiency. MATERIALS AND METHODS The aim of this randomized and blinded study was to assess the effects of NXY-059 on human stem cell-derived neurons in the presence of ischemia-like injury induced by oxygen glucose deprivation or oxidative stress induced by hydrogen peroxide or sodium nitroprusside. RESULTS In MTT assays of cell survival, lactate dehydrogenase assays of total cell death and terminal deoxynucleotidyl transferase dUTP nick end labeling staining of apoptotic-like cell death, NXY-059 at concentrations ranging from 1 µm to 1 mm was completely without activity. Conversely an antioxidant cocktail comprising 100 µm each of ascorbate, reduced glutathione, and dithiothreitol used as a positive control provided marked neuronal protection in these assays. CONCLUSION These findings support our hypothesis that stroke drug screening in human neural tissues will be of value and provides an explanation for the failure of NXY-059 as a human stroke drug.
Collapse
Affiliation(s)
- Ana Antonic
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Victoria, Australia; Department of Neuroscience, Central Clinical School, Monash University, The Alfred Centre, Victoria, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, Centre for Molecular and Medical Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Malcolm R Macleod
- Department of Clinical Neurosciences, University of Edinburgh, Edinburgh, UK
| | - Geoffrey A Donnan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Victoria, Australia
| | - David W Howells
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Victoria, Australia; University of Tasmania, School of Medicine, Faculty of Health, Hobart, Tasmania, Australia.
| |
Collapse
|
58
|
Wright H, Wright T, Pohlig RT, Kasner SE, Raser-Schramm J, Reisman D. Protocol for promoting recovery optimization of walking activity in stroke (PROWALKS): a randomized controlled trial. BMC Neurol 2018; 18:39. [PMID: 29649992 PMCID: PMC5898044 DOI: 10.1186/s12883-018-1044-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Stroke survivors are more physically inactive than even the most sedentary older adults, and low activity is associated with increased risk of recurrent stroke, medical complications, and mortality. We hypothesize that the combination of a fast walking intervention that improves walking capacity, with a step activity monitoring program that facilitates translation of gains from the clinic to the "real-world", would generate greater improvements in real world walking activity than with either intervention alone. METHODS Using a single-blind randomized controlled experimental design, 225 chronic (> 6 months) stroke survivors complete 12 weeks of fast walking training, a step activity monitoring program or a fast walking training + step activity monitoring program. Main eligibility criteria include: chronic ischemic or hemorrhagic stroke (> 6 months post), no evidence of cerebellar stroke, baseline walking speed between 0.3 m/s and 1.0 m/s, and baseline average steps / day < 8000. The primary (steps per day), secondary (self-selected and fastest walking speed, walking endurance, oxygen consumption) and exploratory (vascular events, blood lipids, glucose, blood pressure) outcomes are assessed prior to initiating treatment, after the last treatment and at a 6 and 12-month follow-up. Moderation of the changes in outcomes by baseline characteristics are evaluated to determine for whom the interventions are effective. DISCUSSION Following completion of this study, we will not only understand the efficacy of the interventions and the individuals for which they are effective, we will have the necessary information to design a study investigating the secondary prevention benefits of improved physical activity post-stroke. This study is, therefore, an important step in the development of both rehabilitative and secondary prevention guidelines for persons with stroke. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02835313 . First Posted: July 18, 2016.
Collapse
Affiliation(s)
- Henry Wright
- Department of Physical Therapy, University of Delaware, Newark, DE 19713 USA
| | - Tamara Wright
- Department of Physical Therapy, University of Delaware, Newark, DE 19713 USA
| | - Ryan T. Pohlig
- Biostatistics Core Facility, University of Delaware, Newark, DE 19713 USA
| | - Scott E. Kasner
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | | | - Darcy Reisman
- Department of Physical Therapy, University of Delaware, Newark, DE 19713 USA
| |
Collapse
|
59
|
Muñoz MD, Della Vedova MC, Bushel PR, Ganini da Silva D, Mason RP, Zhai Z, Gomez Mejiba SE, Ramirez DC. The nitrone spin trap 5,5-dimethyl-1-pyrroline N-oxide dampens lipopolysaccharide-induced transcriptomic changes in macrophages. Inflamm Res 2018; 67:515-530. [PMID: 29589052 DOI: 10.1007/s00011-018-1141-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/26/2018] [Accepted: 03/21/2018] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE M1-like inflammatory phenotype of macrophages plays a critical role in tissue damage in chronic inflammatory diseases. Previously, we found that the nitrone spin trap 5,5-dimethyl-1-pyrroline N-oxide (DMPO) dampens lipopolysaccharide (LPS)-triggered inflammatory priming of RAW 264.7 cells. Herein, we tested whether DMPO by itself can induce changes in macrophage transcriptome, and that these effects may prevent LPS-induced activation of macrophages. MATERIALS AND METHODS To test our hypothesis, we performed a transcriptomic and bioinformatics analysis in RAW 264.7 cells incubated with or without LPS, in the presence or in the absence of DMPO. RESULTS Functional data analysis showed 79 differentially expressed genes (DEGs) when comparing DMPO vs Control. We used DAVID databases for identifying enriched gene ontology terms and Ingenuity Pathway Analysis for functional analysis. Our data showed that DMPO vs Control comparison of DEGs is related to downregulation immune-system processes among others. Functional analysis indicated that interferon-response factor 7 and toll-like receptor were related (predicted inhibitions) to the observed transcriptomic effects of DMPO. Functional data analyses of the DMPO + LPS vs LPS DEGs were consistent with DMPO-dampening LPS-induced inflammatory transcriptomic profile in RAW 264.7. These changes were confirmed using Nanostring technology. CONCLUSIONS Taking together our data, surprisingly, indicate that DMPO by itself affects gene expression related to regulation of immune system and that DMPO dampens LPS-triggered MyD88- and TRIF-dependent signaling pathways. Our research provides critical data for further studies on the possible use of DMPO as a structural platform for the design of novel mechanism-based anti-inflammatory drugs.
Collapse
Affiliation(s)
- M D Muñoz
- Laboratory of Experimental and Translational Medicine, IMIBIO-SL-School of Chemistry, Biochemistry and Pharmacy, National University of San Luis-CONICET, San Luis, 5700, San Luis, Argentina.,Laboratory of Experimental Therapeutics, School of Health Sciences-IMIBIO-SL, CONICET-National University of San Luis, San Luis, 5700, San Luis, Argentina
| | - M C Della Vedova
- Laboratory of Experimental and Translational Medicine, IMIBIO-SL-School of Chemistry, Biochemistry and Pharmacy, National University of San Luis-CONICET, San Luis, 5700, San Luis, Argentina.,Laboratory of Experimental Therapeutics, School of Health Sciences-IMIBIO-SL, CONICET-National University of San Luis, San Luis, 5700, San Luis, Argentina
| | - P R Bushel
- Biostatistics and Computational Biology Branch, NIEHS, NIH, USDHHS, RTP, Durham, 27709, NC, USA
| | - D Ganini da Silva
- Immunity, Inflammation and Disease Laboratory, NIEHS, NIH, USDHHS, RTP, Durham, 27709, NC, USA
| | - R P Mason
- Immunity, Inflammation and Disease Laboratory, NIEHS, NIH, USDHHS, RTP, Durham, 27709, NC, USA
| | - Z Zhai
- Department of Dermatology, University of Colorado Denver, Aurora, 80045, CO, USA
| | - S E Gomez Mejiba
- Laboratory of Experimental Therapeutics, School of Health Sciences-IMIBIO-SL, CONICET-National University of San Luis, San Luis, 5700, San Luis, Argentina.
| | - D C Ramirez
- Laboratory of Experimental and Translational Medicine, IMIBIO-SL-School of Chemistry, Biochemistry and Pharmacy, National University of San Luis-CONICET, San Luis, 5700, San Luis, Argentina.
| |
Collapse
|
60
|
Yang J, Qi J, Xiu B, Yang B, Niu C, Yang H. Reactive Oxygen Species Play a Biphasic Role in Brain Ischemia. J INVEST SURG 2018; 32:97-102. [PMID: 29420085 DOI: 10.1080/08941939.2017.1376131] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Jiping Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jinchong Qi
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Baoxin Xiu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Bei Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Caihong Niu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Hua Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
61
|
Neuroprotective Effect and Mechanism of Action of Tetramethylpyrazine Nitrone for Ischemic Stroke Therapy. Neuromolecular Med 2018; 20:97-111. [DOI: 10.1007/s12017-018-8478-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/18/2018] [Indexed: 10/18/2022]
|
62
|
Aicardo A, Mastrogiovanni M, Cassina A, Radi R. Propagation of free-radical reactions in concentrated protein solutions. Free Radic Res 2018; 52:159-170. [DOI: 10.1080/10715762.2017.1420905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Adrián Aicardo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Mauricio Mastrogiovanni
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Adriana Cassina
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
63
|
Savitz SI, Baron JC, Yenari MA, Sanossian N, Fisher M. Reconsidering Neuroprotection in the Reperfusion Era. Stroke 2017; 48:3413-3419. [PMID: 29146878 DOI: 10.1161/strokeaha.117.017283] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/18/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Sean I Savitz
- From the Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.); Department of Neurology, UTHealth, Houston, TX (S.I.S.); Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France (J.-C.B.); Department of Neurology, University of California, San Francisco (M.A.Y.); Department of Neurology, San Francisco VA Medical Center, CA (M.A.Y.); Roxanna Todd Hodges Comprehensive Stroke Clinic, Los Angeles, CA (N.S.); Department of Neurology, University of Southern California, Los Angeles (N.S.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.).
| | - Jean-Claude Baron
- From the Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.); Department of Neurology, UTHealth, Houston, TX (S.I.S.); Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France (J.-C.B.); Department of Neurology, University of California, San Francisco (M.A.Y.); Department of Neurology, San Francisco VA Medical Center, CA (M.A.Y.); Roxanna Todd Hodges Comprehensive Stroke Clinic, Los Angeles, CA (N.S.); Department of Neurology, University of Southern California, Los Angeles (N.S.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Midori A Yenari
- From the Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.); Department of Neurology, UTHealth, Houston, TX (S.I.S.); Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France (J.-C.B.); Department of Neurology, University of California, San Francisco (M.A.Y.); Department of Neurology, San Francisco VA Medical Center, CA (M.A.Y.); Roxanna Todd Hodges Comprehensive Stroke Clinic, Los Angeles, CA (N.S.); Department of Neurology, University of Southern California, Los Angeles (N.S.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Nerses Sanossian
- From the Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.); Department of Neurology, UTHealth, Houston, TX (S.I.S.); Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France (J.-C.B.); Department of Neurology, University of California, San Francisco (M.A.Y.); Department of Neurology, San Francisco VA Medical Center, CA (M.A.Y.); Roxanna Todd Hodges Comprehensive Stroke Clinic, Los Angeles, CA (N.S.); Department of Neurology, University of Southern California, Los Angeles (N.S.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Marc Fisher
- From the Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.); Department of Neurology, UTHealth, Houston, TX (S.I.S.); Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France (J.-C.B.); Department of Neurology, University of California, San Francisco (M.A.Y.); Department of Neurology, San Francisco VA Medical Center, CA (M.A.Y.); Roxanna Todd Hodges Comprehensive Stroke Clinic, Los Angeles, CA (N.S.); Department of Neurology, University of Southern California, Los Angeles (N.S.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| |
Collapse
|
64
|
Constitutive Expression of Adiponectin in Endothelial Progenitor Cells Protects a Rat Model of Cerebral Ischemia. Neural Plast 2017; 2017:6809745. [PMID: 29201467 PMCID: PMC5671740 DOI: 10.1155/2017/6809745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/15/2017] [Indexed: 02/05/2023] Open
Abstract
Endothelial progenitor cells (EPCs), as precursors to endothelial cells, play a significant part in the process of endogenous blood vessel repair and maintenance of endothelial integrity. Adiponectin (APN) is an adipocyte-specific adipocytokine. In this study, we aim to test whether we transplant a combined graft of EPCs transfected with the adiponectin gene into a rat model of cerebral ischemia could improve functional recovery after middle cerebral artery occlusion (MCAO). Sprague-Dawley (SD) rats were randomly divided into a MCAO control group, a MCAO EPC treatment group, and a MCAO LV-APN-EPC treatment group. A focal cerebral ischemia and reperfusion model was induced by the intraluminal suture method. After 2 h of reperfusion, EPCs were transplanted by injection through the tail vein. A rotarod test was conducted to assess behavioral function before MCAO and on days 1, 7, and 14 after MCAO. After 14 d, TTC staining, CD31 immunofluorescence, and TUNEL staining were used to evaluate infarct volume, microvessel density, and cell apoptosis. Results revealed that behavioral function, infarct area percentage, microvessel density, and cell apoptosis rates were more favorable in the LV-APN-EPC treatment group than in the EPC treatment group. These data suggested that gene-modified cell therapy may be a useful approach for the treatment of ischemic stroke.
Collapse
|
65
|
Ayuso MI, Martínez-Alonso E, Chioua M, Escobar-Peso A, Gonzalo-Gobernado R, Montaner J, Marco-Contelles J, Alcázar A. Quinolinyl Nitrone RP19 Induces Neuroprotection after Transient Brain Ischemia. ACS Chem Neurosci 2017; 8:2202-2213. [PMID: 28731692 DOI: 10.1021/acschemneuro.7b00126] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
There is a need to develop additional effective therapies for ischemic stroke. Nitrones, which were first developed as reactive oxygen species (ROS)-trapping compounds, have been proposed as neuroprotective agents for ischemic stroke, a ROS-related disorder. The previous reported ROS-trapping compound, quinolyl nitrone RP19, is here being assayed to induce neuroprotection to ischemia-reperfusion injury in three experimental ischemia models: (i) oxygen-glucose deprivation (OGD) on primary neuronal cultures; (ii) transient global cerebral ischemia in four-vessel occlusion model; and (iii) transient focal cerebral ischemia in middle cerebral artery occlusion (tMCAO) model. RP19 (50 μM) induced long-term neuroprotection at 5 days of recovery after OGD in primary neuronal cultures, evaluated by cell viability assay, and decreased both ROS formation and lipid peroxidation upon recovery after OGD. Furthermore, treatment of animals with RP19 at the onset of reperfusion after either global or focal ischemia, at the dose range that was demonstrated to be neuroprotective in neuronal cultures, decreased neuronal death and apoptosis induction, reduced the size of infarct, and improved the neurological deficit scores after 48 h or 5 days of reperfusion after ischemia. The molecule proposed, quinolyl nitrone RP19, induced substantial neuroprotection on experimental ischemia in neuronal cells, and against ischemic injury following transient brain ischemia in treated animals. This molecule may have potential therapeutic interest in ischemic stroke and to reduce the reoxygenation-induced injury after induced reperfusion.
Collapse
Affiliation(s)
- Maria I. Ayuso
- Department of Investigation, Hospital Ramón y Cajal, IRYCIS, Madrid 28034, Spain
- Neurovascular Research Laboratory, Institut
de Recerca Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona 08035, Spain
- Neurovascular
Research Group, Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, Sevilla 41013, Spain
| | - Emma Martínez-Alonso
- Department of Investigation, Hospital Ramón y Cajal, IRYCIS, Madrid 28034, Spain
| | - Mourad Chioua
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC), Madrid 28006, Spain
| | - Alejandro Escobar-Peso
- Department of Investigation, Hospital Ramón y Cajal, IRYCIS, Madrid 28034, Spain
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC), Madrid 28006, Spain
| | - Rafael Gonzalo-Gobernado
- Neurovascular
Research Group, Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, Sevilla 41013, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Institut
de Recerca Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona 08035, Spain
- Neurovascular
Research Group, Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, Sevilla 41013, Spain
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC), Madrid 28006, Spain
| | - Alberto Alcázar
- Department of Investigation, Hospital Ramón y Cajal, IRYCIS, Madrid 28034, Spain
| |
Collapse
|
66
|
HPN-07, a free radical spin trapping agent, protects against functional, cellular and electrophysiological changes in the cochlea induced by acute acoustic trauma. PLoS One 2017; 12:e0183089. [PMID: 28832600 PMCID: PMC5568441 DOI: 10.1371/journal.pone.0183089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/28/2017] [Indexed: 11/29/2022] Open
Abstract
Oxidative stress is considered a major cause of the structural and functional changes associated with auditory pathologies induced by exposure to acute acoustic trauma AAT). In the present study, we examined the otoprotective effects of 2,4-disulfophenyl-N-tert-butylnitrone (HPN-07), a nitrone-based free radical trap, on the physiological and cellular changes in the auditory system of chinchilla following a six-hour exposure to 4 kHz octave band noise at 105 dB SPL. HPN-07 has been shown to suppress oxidative stress in biological models of a variety of disorders. Our results show that administration of HPN-07 beginning four hours after acoustic trauma accelerated and enhanced auditory/cochlear functional recovery, as measured by auditory brainstem responses (ABR), distortion product otoacoustic emissions (DPOAE), compound action potentials (CAP), and cochlear microphonics (CM). The normally tight correlation between the endocochlear potential (EP) and evoked potentials of CAP and CM were persistently disrupted after noise trauma in untreated animals but returned to homeostatic conditions in HPN-07 treated animals. Histological analyses revealed several therapeutic advantages associated with HPN-07 treatment following AAT, including reductions in inner and outer hair cell loss; reductions in AAT-induced loss of calretinin-positive afferent nerve fibers in the spiral lamina; and reductions in fibrocyte loss within the spiral ligament. These findings support the conclusion that early intervention with HPN-07 following an AAT efficiently blocks the propagative ototoxic effects of oxidative stress, thereby preserving the homeostatic and functional integrity of the cochlea.
Collapse
|
67
|
Babadjouni RM, Radwanski RE, Walcott BP, Patel A, Durazo R, Hodis DM, Emanuel BA, Mack WJ. Neuroprotective strategies following intraparenchymal hemorrhage. J Neurointerv Surg 2017; 9:1202-1207. [PMID: 28710084 DOI: 10.1136/neurintsurg-2017-013197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/23/2022]
Abstract
Intracerebral hemorrhage and, more specifically, intraparenchymal hemorrhage, are devastating disease processes with poor clinical outcomes. Primary injury to the brain results from initial hematoma expansion while secondary hemorrhagic injury occurs from blood-derived products such as hemoglobin, heme, iron, and coagulation factors that overwhelm the brains natural defenses. Novel neuroprotective treatments have emerged that target primary and secondary mechanisms of injury. Nonetheless, translational application of neuroprotectants from preclinical to clinical studies has yet to show beneficial clinical outcomes. This review summarizes therapeutic agents and neuroprotectants in ongoing clinical trials aimed at targeting primary and secondary mechanisms of injury after intraparenchymal hemorrhage.
Collapse
Affiliation(s)
- Robin Moshe Babadjouni
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ryan E Radwanski
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Brian P Walcott
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Arati Patel
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ramon Durazo
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Drew M Hodis
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Benjamin A Emanuel
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - William J Mack
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
68
|
Regenhardt RW, Das AS, Stapleton CJ, Chandra RV, Rabinov JD, Patel AB, Hirsch JA, Leslie-Mazwi TM. Blood Pressure and Penumbral Sustenance in Stroke from Large Vessel Occlusion. Front Neurol 2017; 8:317. [PMID: 28717354 PMCID: PMC5494536 DOI: 10.3389/fneur.2017.00317] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022] Open
Abstract
The global burden of stroke remains high, and of the various subtypes of stroke, large vessel occlusions (LVOs) account for the largest proportion of stroke-related death and disability. Several randomized controlled trials in 2015 changed the landscape of stroke care worldwide, with endovascular thrombectomy (ET) now the standard of care for all eligible patients. With the proven success of this therapy, there is a renewed focus on penumbral sustenance. In this review, we describe the ischemic penumbra, collateral circulation, autoregulation, and imaging assessment of the penumbra. Blood pressure goals in acute stroke remain controversial, and we review the current data and suggest an approach for induced hypertension in the acute treatment of patients with LVOs. Finally, in addition to reperfusion and enhanced perfusion, efforts focused on developing therapeutic targets that afford neuroprotection and augment neural repair will gain increasing importance. ET has revolutionized stroke care, and future emphasis will be placed on promoting penumbral sustenance, which will increase patient eligibility for this highly effective therapy and reduce overall stroke-related death and disability.
Collapse
Affiliation(s)
- Robert W. Regenhardt
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Alvin S. Das
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Christopher J. Stapleton
- Neuroendovascular Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Ronil V. Chandra
- Interventional Neuroradiology, Monash Imaging, Monash Health, Monash University, Melbourne, VIC, Australia
| | - James D. Rabinov
- Neuroendovascular Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Aman B. Patel
- Neuroendovascular Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua A. Hirsch
- Neuroendovascular Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Thabele M. Leslie-Mazwi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Neuroendovascular Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
69
|
Affiliation(s)
- Joseph P Broderick
- From the Departments of Neurology and Rehabilitation Medicine (J.P.B.) and Emergency Medicine (O.A.), University of Cincinnati Gardner Neuroscience Institute, University of Cincinnati, OH; and Division of Biostatistics, Medical University of South Carolina, Charleston (J.E.).
| | - Opeolu Adeoye
- From the Departments of Neurology and Rehabilitation Medicine (J.P.B.) and Emergency Medicine (O.A.), University of Cincinnati Gardner Neuroscience Institute, University of Cincinnati, OH; and Division of Biostatistics, Medical University of South Carolina, Charleston (J.E.)
| | - Jordan Elm
- From the Departments of Neurology and Rehabilitation Medicine (J.P.B.) and Emergency Medicine (O.A.), University of Cincinnati Gardner Neuroscience Institute, University of Cincinnati, OH; and Division of Biostatistics, Medical University of South Carolina, Charleston (J.E.)
| |
Collapse
|
70
|
Sifat AE, Vaidya B, Abbruscato TJ. Blood-Brain Barrier Protection as a Therapeutic Strategy for Acute Ischemic Stroke. AAPS JOURNAL 2017; 19:957-972. [PMID: 28484963 DOI: 10.1208/s12248-017-0091-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/18/2017] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) is a vital component of the neurovascular unit (NVU) containing tight junctional (TJ) proteins and different ion and nutrient transporters which maintain normal brain physiology. BBB disruption is a major pathological hallmark in the course of ischemic stroke which is regulated by the actions of different factors working at different stages of cerebral ischemia including matrix metalloproteinases (MMPs), inflammatory modulators, vesicular trafficking, oxidative pathways, and junctional-cytoskeletal interactions. These components interact further to disrupt maintenance of both the paracellular and transport barriers of the central nervous system (CNS) to worsen ischemic brain injury and the propensity for hemorrhagic transformation (HT) associated with injury and/or thrombolytic therapy with tissue-type plasminogen activator (tPA). We propose that these complex molecular pathways should be evaluated further so that they could be targeted alone or in combination to protect the BBB during cerebral ischemia. These types of novel interventions should be guided by advanced imaging techniques for better diagnosis of BBB damage which may exert significant therapeutic benefit including the extension of therapeutic window of tPA. This review will focus on the different stages and mechanisms of BBB damage in acute ischemic stroke and novel therapeutic strategies to target those pathways for better therapeutic outcome in stroke.
Collapse
Affiliation(s)
- Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter, Amarillo, Texas, 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter, Amarillo, Texas, 79106, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter, Amarillo, Texas, 79106, USA.
| |
Collapse
|
71
|
Karsy M, Brock A, Guan J, Taussky P, Kalani MYS, Park MS. Neuroprotective strategies and the underlying molecular basis of cerebrovascular stroke. Neurosurg Focus 2017; 42:E3. [DOI: 10.3171/2017.1.focus16522] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Stroke is a leading cause of disability in the US. Although there has been significant progress in the area of medical and surgical thrombolytic technologies, neuroprotective agents to prevent secondary cerebral injury and to minimize disability remain limited. Only limited success has been reported in preclinical and clinical trials evaluating a variety of compounds. In this review, the authors discuss the most up-to-date information regarding the underlying molecular biology of stroke as well as strategies that aim to mitigate this complex signaling cascade. Results of historical research trials involving N-methyl-d-aspartate and α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor antagonists, clomethiazole, antioxidants, citicoline, nitric oxide, and immune regulators have laid the groundwork for current progress. In addition, more recent studies involving therapeutic hypothermia, magnesium, albumin, glyburide, uric acid, and a variety of other treatments have provided more options. The use of neuroprotective agents in combination or with existing thrombolytic treatments may be one of many exciting areas of further development. Although past trials of neuroprotective agents in ischemic stroke have been limited, significant insights into mechanisms of stroke, animal models, and trial design have incrementally improved approaches for future therapies.
Collapse
|
72
|
Stroke Cytoprotection: Can Repeating History with New Expectations Really Be the Path to Success in Stroke Research? Transl Stroke Res 2017; 8:104-106. [DOI: 10.1007/s12975-017-0528-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 02/16/2017] [Indexed: 01/05/2023]
|
73
|
|
74
|
Escobar-Peso A, Chioua M, Frezza V, Martínez-Alonso E, Marco-Contelles J, Alcázar A. Nitrones, Old Fellows for New Therapies in Ischemic Stroke. SPRINGER SERIES IN TRANSLATIONAL STROKE RESEARCH 2017. [DOI: 10.1007/978-3-319-45345-3_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
75
|
Li W, Yang S. Targeting oxidative stress for the treatment of ischemic stroke: Upstream and downstream therapeutic strategies. Brain Circ 2016; 2:153-163. [PMID: 30276293 PMCID: PMC6126224 DOI: 10.4103/2394-8108.195279] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/04/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022] Open
Abstract
Excessive oxygen and its chemical derivatives, namely reactive oxygen species (ROS), produce oxidative stress that has been known to lead to cell injury in ischemic stroke. ROS can damage macromolecules such as proteins and lipids and leads to cell autophagy, apoptosis, and necrosis to the cells. This review describes studies on the generation of ROS, its role in the pathogenesis of ischemic stroke, and recent development in therapeutic strategies in reducing oxidative stress after ischemic stroke.
Collapse
Affiliation(s)
- Wenjun Li
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shaohua Yang
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
76
|
Fong JJ, Rhoney DH. NXY-059: Review of Neuroprotective Potential for Acute Stroke. Ann Pharmacother 2016; 40:461-71. [PMID: 16507608 DOI: 10.1345/aph.1e636] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Objective: To review available literature on the pharmacology, pharmacokinetics, efficacy, and tolerability of NXY-059, an Investigational agent with a potential role in the treatment of acute stroke. Data Sources: Information was obtained from a MEDLINE search (1966–February 2006) of English-language literature utilizing the following search terms: NXY-059, cerovive, nitrones, neuroprotection, free radical trapper, and secondary neurologic injury. Study Selection and Data Extraction: Data from animal and human trials were evaluated to summarize the mechanism of action, efficacy, and safety of NXY-059. All published and unpublished trials and abstracts citing NXY-059 were selected. Data Synthesis: NXY-059 is an intravenous, nitrone-based, free radical trapping agent in Phase III trials for treatment of acute stroke. In various animal models, NXY-059 has shown reductions in infarct volume and neurologic deficits. Pharmacokinetic studies indicate that NXY-059 displays a predictable pharmacokinetic profile and primarily undergoes renal elimination. Results from 2 Phase II clinical trials showed favorable results for the safety and tolerability of the drug. A recent analysis of one of the Phase III trials showed a statistically significant reduction in the primary outcome of disability after acute stroke in patients who received NXY-059 compared with placebo. Conclusions: NXY-059 is a novel agent undergoing worldwide Phase III trials. Initial safety and efficacy data have not revealed any serious adverse events requiring special monitoring and/or precautions, with the exception of drug accumulation in patients with renal insufficiency. The potential benefit of this agent can change the current management algorithm for acute stroke and may represent significant advancement for the care of these patients.
Collapse
Affiliation(s)
- Jeffrey J Fong
- Tufts-New England Medical Center, Northeastern University School of Pharmacy, Boston, MA, USA
| | | |
Collapse
|
77
|
Yao Y, Miao W, Liu Z, Han W, Shi K, Shen Y, Li H, Liu Q, Fu Y, Huang D, Shi FD. Dimethyl Fumarate and Monomethyl Fumarate Promote Post-Ischemic Recovery in Mice. Transl Stroke Res 2016; 7:535-547. [PMID: 27614618 PMCID: PMC5065588 DOI: 10.1007/s12975-016-0496-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 01/21/2023]
Abstract
Oxidative stress plays an important role in cerebral ischemia-reperfusion injury. Dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) are antioxidant agents that can activate the nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway and induce the expression of antioxidant proteins. Here, we evaluated the impact of DMF and MMF on ischemia-induced brain injury and whether the Nrf2 pathway mediates the effects provided by DMF and MMF in cerebral ischemia-reperfusion injury. Using a mouse model of transient focal brain ischemia, we show that DMF and MMF significantly reduce neurological deficits, infarct volume, brain edema, and cell death. Further, DMF and MMF suppress glial activation following brain ischemia. Importantly, the protection of DMF and MMF was mostly evident during the subacute stage and was abolished in Nrf2-/- mice, indicating that the Nrf2 pathway is required for the beneficial effects of DMF and MMF. Together, our data indicate that DMF and MMF have therapeutic potential in cerebral ischemia-reperfusion injury and their protective role is likely mediated by the Nrf2 pathway.
Collapse
Affiliation(s)
- Yang Yao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Weimin Miao
- The State Key Laboratory of Experimental Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Zhijia Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wei Han
- Department of Radiology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Kaibin Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yi Shen
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Handong Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Ying Fu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - DeRen Huang
- Neurology and Neuroscience Associates, Unity Health Network, Akron, OH, USA
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA.
| |
Collapse
|
78
|
Tetramethylpyrazine nitrone, a multifunctional neuroprotective agent for ischemic stroke therapy. Sci Rep 2016; 6:37148. [PMID: 27841332 PMCID: PMC5107909 DOI: 10.1038/srep37148] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/25/2016] [Indexed: 11/08/2022] Open
Abstract
TBN, a novel tetramethylpyrazine derivative armed with a powerful free radical-scavenging nitrone moiety, has been reported to reduce cerebral infarction in rats through multi-functional mechanisms of action. Here we study the therapeutic effects of TBN on non-human primate model of stroke. Thirty male Cynomolgus macaques were subjected to stroke with 4 hours ischemia and then reperfusion. TBN were injected intravenously at 3 or 6 hours after the onset of ischemia. Cerebral infarction was examined by magnetic resonance imaging at 1 and 4 weeks post ischemia. Neurological severity scores were evaluated during 4 weeks observation. At the end of experiment, protein markers associated with the stroke injury and TBN treatment were screened by quantitative proteomics. We found that TBN readily penetrated the blood brain barrier and reached effective therapeutic concentration after intravenous administration. It significantly reduced brain infarction and modestly preserved the neurological function of stroke-affected arm. TBN suppressed over-expression of neuroinflammatory marker vimentin and decreased the numbers of GFAP-positive cells, while reversed down-regulation of myelination-associated protein 2', 3'-cyclic-nucleotide 3'-phosphodiesterase and increased the numbers of NeuN-positive cells in the ipsilateral peri-infarct area. TBN may serve as a promising new clinical candidate for the treatment of ischemic stroke.
Collapse
|
79
|
Ginsberg MD. Expanding the concept of neuroprotection for acute ischemic stroke: The pivotal roles of reperfusion and the collateral circulation. Prog Neurobiol 2016; 145-146:46-77. [PMID: 27637159 DOI: 10.1016/j.pneurobio.2016.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/22/2016] [Accepted: 09/10/2016] [Indexed: 12/27/2022]
Abstract
This review surveys the efforts taken to achieve clinically efficacious protection of the ischemic brain and underscores the necessity of expanding our purview to include the essential role of cerebral perfusion and the collateral circulation. We consider the development of quantitative strategies to measure cerebral perfusion at the regional and local levels and the application of these methods to elucidate flow-related thresholds of ischemic viability and to characterize the ischemic penumbra. We stress that the modern concept of neuroprotection must consider perfusion, the necessary substrate upon which ischemic brain survival depends. We survey the major mechanistic approaches to neuroprotection and review clinical neuroprotection trials, focusing on those phase 3 multicenter clinical trials for acute ischemic stroke that have been completed or terminated. We review the evolution of thrombolytic therapies; consider the lessons learned from the initial, negative multicenter trials of endovascular therapy; and emphasize the highly successful positive trials that have finally established a clinical role for endovascular clot removal. As these studies point to the brain's collateral circulation as key to successful reperfusion, we next review the anatomy and pathophysiology of collateral perfusion as it relates to ischemic infarction, as well as the molecular and genetic influences on collateral development. We discuss the current MR and CT-based diagnostic methods for assessing the collateral circulation and the prognostic significance of collaterals in ischemic stroke, and we consider past and possible future therapeutic directions.
Collapse
Affiliation(s)
- Myron D Ginsberg
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
80
|
Affiliation(s)
- Stuart J Pocock
- From the Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London (S.J.P.); and Columbia University Medical Center, New York Presbyterian Hospital, and the Cardiovascular Research Foundation - all in New York (G.W.S.)
| | - Gregg W Stone
- From the Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London (S.J.P.); and Columbia University Medical Center, New York Presbyterian Hospital, and the Cardiovascular Research Foundation - all in New York (G.W.S.)
| |
Collapse
|
81
|
Kent TA, Mandava P. Embracing Biological and Methodological Variance in a New Approach to Pre-Clinical Stroke Testing. Transl Stroke Res 2016; 7:274-83. [PMID: 27018014 PMCID: PMC5425098 DOI: 10.1007/s12975-016-0463-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/08/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Abstract
High-profile failures in stroke clinical trials have discouraged clinical translation of neuroprotectants. While there are several plausible explanations for these failures, we believe that the fundamental problem is the way clinical and pre-clinical studies are designed and analyzed for heterogeneous disorders such as stroke due to innate biological and methodological variability that current methods cannot capture. Recent efforts to address pre-clinical rigor and design, while important, are unable to account for variability present even in genetically homogenous rodents. Indeed, efforts to minimize variability may lessen the clinical relevance of pre-clinical models. We propose a new approach that recognizes the important role of baseline stroke severity and other factors in influencing outcome. Analogous to clinical trials, we propose reporting baseline factors that influence outcome and then adapting for the pre-clinical setting a method developed for clinical trial analysis where the influence of baseline factors is mathematically modeled and the variance quantified. A new therapy's effectiveness is then evaluated relative to the pooled outcome variance at its own baseline conditions. In this way, an objective threshold for robustness can be established that must be overcome to suggest its effectiveness when expanded to broader populations outside of the controlled environment of the PI's laboratory. The method is model neutral and subsumes sources of variance as reflected in baseline factors such as initial stroke severity. We propose that this new approach deserves consideration for providing an objective method to select agents worthy of the commitment of time and resources in translation to clinical trials.
Collapse
Affiliation(s)
- Thomas A Kent
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, McNair Campus, 7200 Cambridge St. 9th Floor, MS: BCM609, Houston, TX, 77030, USA.
- Michael E. DeBakey VA Medical Center Stroke Program and Center for Translational Research on Inflammatory Diseases, Houston, TX, USA.
| | - Pitchaiah Mandava
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, McNair Campus, 7200 Cambridge St. 9th Floor, MS: BCM609, Houston, TX, 77030, USA
- Michael E. DeBakey VA Medical Center Stroke Program and Center for Translational Research on Inflammatory Diseases, Houston, TX, USA
| |
Collapse
|
82
|
Chamorro Á, Dirnagl U, Urra X, Planas AM. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol 2016; 15:869-881. [DOI: 10.1016/s1474-4422(16)00114-9] [Citation(s) in RCA: 556] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/15/2016] [Accepted: 03/03/2016] [Indexed: 01/04/2023]
|
83
|
|
84
|
Abstract
Background: Measures of damage limitation for acute stroke have not produced substantial benefit to reduce stroke mortality. Search continues for measures to reduce stroke mortality. Methods: Literature review for influence of cardiovascular factors, specifically the value of NT proBNP (a sensitive index of cardiac impairment) for stroke mortality. Results: Cardiovascular factors, in particular cardiac failure, adversely influence acute stroke mortality. There is evidence of cardiological abnormality in acute strokes as indicated by ECG changes and tachycardia secondary to neurohumoral changes in acute strokes. Patients with ECG abnormality, tachycardia, dysrrhythmia and elevated levels of nor-epi-nephrine in acute stroke phase have higher mortality. Recent studies reveal that Troponin (measure of cardiac injury) and NT-proBNP (measure of cardiac function impairment) are elevated in acute stroke patients, in response to the activated Renin-Angiotensin-Aldosterone-System and other neurohumoral changes, as a protective mechanism for sympathoinhibitory activity. Patients with elevated troponin have a higher mortality. Similarly elevated NT-proBNP has been reported to be associated with higher short and long-term mortality. In one study all patients who died at 4 months had NT-proBNP levels above the median, no patient with NT-proBNP below the median value died. Two studies revealed that NT-proBNP is more significant than clinical stroke severity for stroke mortality. Protection of myocardium in stroke patients may be possible by the use of drugs such as beta-blockers and the drugs acting on RAAS. Reduction of mortality in studies of candesartan (ACCESS study) and prior beta-blockers is one such example. Heart is at risk in acute strokes and protecting heart makes sense to reduce stroke mortality. Conclusion: Some stroke patients die due to occult cardiac damage and functional impairment in acute phase due to common risk factors. This relationship between brain and heart needs evaluation. Protection of heart with currently available or new drugs in acute strokes is worth investigating since this intervention could be applied to a large proportion of acute stroke patients over a wide time window.
Collapse
Affiliation(s)
| | - Ian Ross
- Kings Mill Hospital, Sutton in Ashfield, Notts NG17 4JL, UK
| | | |
Collapse
|
85
|
Affiliation(s)
| | - Ian Ross
- Kings Mill Hospital, Sutton in Ashfield, Notts, UK
| | | |
Collapse
|
86
|
|
87
|
Weber J, Ebinger M, Audebert HJ. Prehospital stroke care: telemedicine, thrombolysis and neuroprotection. Expert Rev Neurother 2016; 15:753-61. [PMID: 26109228 DOI: 10.1586/14737175.2015.1051967] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Over the last 15 years, new approaches regarding neuroprotective and thrombolytic strategies in stroke management have been evaluated in the prehospital setting. These efforts have provided exciting new potentials of hyperacute stroke care. Trials have shown that the use of specialized stroke ambulances increases the proportion of patients receiving intravenous thrombolysis and shortens alarm-to-treatment time by approximately half an hour compared to standard care. Intravenous thrombolysis within the ultra-early time window of the 'golden hour' has become a realistic scenario. However, direct effects of prehospital stroke care on functional outcome have yet to be shown and other approaches such as neuroprotective treatments could not demonstrate clinical benefit so far. There is a clear need for systematic research in the prehospital field to test the clinical effectiveness and cost-effectiveness of new therapeutic strategies. It will be necessary to test various components of prehospital stroke care alone and in combination.
Collapse
Affiliation(s)
- Joachim Weber
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | |
Collapse
|
88
|
Gonzales NR, Grotta JC. Pharmacologic Modification of Acute Cerebral Ischemia. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00055-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
89
|
|
90
|
Intravenous Thrombolysis. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00051-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
|
91
|
Oxidative Stress in Intracerebral Hemorrhage: Sources, Mechanisms, and Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3215391. [PMID: 26843907 PMCID: PMC4710930 DOI: 10.1155/2016/3215391] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is associated with the highest mortality and morbidity despite only constituting approximately 10–15% of all strokes. Complex underlying mechanisms consisting of cytotoxic, excitotoxic, and inflammatory effects of intraparenchymal blood are responsible for its highly damaging effects. Oxidative stress (OS) also plays an important role in brain injury after ICH but attracts less attention than other factors. Increasing evidence has demonstrated that the metabolite axis of hemoglobin-heme-iron is the key contributor to oxidative brain damage after ICH, although other factors, such as neuroinflammation and prooxidases, are involved. This review will discuss the sources, possible molecular mechanisms, and potential therapeutic targets of OS in ICH.
Collapse
|
92
|
Serum pentraxin-3 levels in acute stroke: No association with stroke prognosis. Atherosclerosis 2015; 243:616-20. [PMID: 26546709 DOI: 10.1016/j.atherosclerosis.2015.10.089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/10/2015] [Accepted: 10/20/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Stroke is the leading cause of serious disability. Estimating severity of the disease and early risk assessment is crucial. Several studies have been carried on and several biomarkers have been proposed in the literature for risk assessment and to estimate the stroke prognosis. In this study we assessed the association of predictors such as patient age, gender, stroke volume and NIHS scores on prognosis of stroke event. We investigated whether the serum pentraxin-3 levels are linked with stroke prognosis. METHODS Forty-four stroke patients without cardiovascular risk factors were included in this study. Initial NIHS scores, stroke volumes, serum pentraxin-3 levels and the data regarding the risk factors were collected in the first and seventh days of event. Association of predictors with final NIHS scores were investigated using multivariate regression model. RESULTS Initial NIHS score, initial and final stroke volumes were independently associated with final NIHS score whereas serum pentraxin-3 levels, whether acquired at the first or seventh day of stroke, were not associated with final NIHS score. CONCLUSIONS In stroke patients without cardiovascular, cardiopulmonary and infectious diseases, serum pentraxin-3 levels are not associated with stroke prognosis.
Collapse
|
93
|
Mandava P, Shah SD, Sarma AK, Kent TA. An Outcome Model for Intravenous rt-PA in Acute Ischemic Stroke. Transl Stroke Res 2015; 6:451-7. [PMID: 26385545 DOI: 10.1007/s12975-015-0427-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 09/04/2015] [Indexed: 01/19/2023]
Abstract
Most early phase trials in stroke and brain trauma have failed in phase 3, including efforts to improve acute ischemic stroke outcomes beyond that achieved by intravenous recombinant tissue plasminogen activator (t-PA) (IVT). With the exception of more recent stent retriever trials, most subsequent phase 3 trials failed. We previously showed that baseline imbalances, non-linear relationships of these factors to outcome, and unrepresentative control populations invalidate traditional statistical analysis in early trials of heterogeneous diseases such as stroke. We developed an alternative approach using a pooled outcome model derived from control arms of randomized clinical trial (RCTs). This model then permits comparing treatment trials to an expected outcome of a pooled population. Here, we hypothesized we could develop such a model for IVT and tested it against outcomes without IVT. We surveyed literature for all trials involving one arm with IVT reporting baseline National Institute Stroke Scale (NIHSS), age, and outcome. A non-linear fit was performed including multi-dimensional statistical intervals (±95 %) permitting visual comparison of outcomes at their own baselines. We compared models derived from non-IVT control arms. Models from 24 IVT RCTs representing 3195 subjects were successfully generated for functional outcome, modified Rankin Scale (mRS) 0-2 (r(2) = 0. 83, p < 0.001), and mortality (r(2) = 0.54; p = 0.001). We confirmed better outcomes compared to no IVT and mixed use IVT models across the range of baseline factors. It was possible to generate an expected outcome model for IVT from existing literature. We confirmed benefit compared to placebo. This model should be useful to compare to new agents without the need for statistical manipulation.
Collapse
Affiliation(s)
- Pitchaiah Mandava
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd (127), Houston, TX, 77030, USA.
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA.
| | - Shreyansh D Shah
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Anand K Sarma
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Thomas A Kent
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd (127), Houston, TX, 77030, USA
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
94
|
van Meer PJ, Graham ML, Schuurman HJ. The safety, efficacy and regulatory triangle in drug development: Impact for animal models and the use of animals. Eur J Pharmacol 2015; 759:3-13. [DOI: 10.1016/j.ejphar.2015.02.055] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/15/2015] [Accepted: 02/09/2015] [Indexed: 11/26/2022]
|
95
|
Kent TA, Shah SD, Mandava P. Improving early clinical trial phase identification of promising therapeutics. Neurology 2015; 85:274-83. [PMID: 26109712 DOI: 10.1212/wnl.0000000000001757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 01/23/2015] [Indexed: 11/15/2022] Open
Abstract
This review addresses decision-making underlying the frequent failure to confirm early-phase positive trial results and how to prioritize which early agents to transition to late phase. While unexpected toxicity is sometimes responsible for late-phase failures, lack of efficacy is also frequently found. In stroke as in other conditions, early trials often demonstrate imbalances in factors influencing outcome. Other issues complicate early trial analysis, including unequally distributed noise inherent in outcome measures and variations in natural history among studies. We contend that statistical approaches to correct for imbalances and noise, while likely valid for homogeneous conditions, appear unable to accommodate disease complexity and have failed to correctly identify effective agents. While blinding and randomization are important to reduce selection bias, these methods appear insufficient to insure valid conclusions. We found potential sources of analytical errors in nearly 90% of a sample of early stroke trials. To address these issues, we recommend changes in early-phase analysis and reporting: (1) restrict use of statistical correction to studies where the underlying assumptions are validated, (2) select dichotomous over continuous outcomes for small samples, (3) consider pooled samples to model natural history to detect early therapeutic signals and increase the likelihood of replication in larger samples, (4) report subgroup baseline conditions, (5) consider post hoc methods to restrict analysis to subjects with an appropriate match, and (6) increase the strength of effect threshold given these cumulative sources of noise and potential errors. More attention to these issues should lead to better decision-making regarding selection of agents to proceed to pivotal trials.
Collapse
Affiliation(s)
- Thomas A Kent
- From The Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine; and Center of Translational Research on Inflammatory Diseases, Michael E. DeBakey Stroke Program, Michael E. DeBakey VA Medical Center, Houston, TX.
| | - Shreyansh D Shah
- From The Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine; and Center of Translational Research on Inflammatory Diseases, Michael E. DeBakey Stroke Program, Michael E. DeBakey VA Medical Center, Houston, TX
| | - Pitchaiah Mandava
- From The Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine; and Center of Translational Research on Inflammatory Diseases, Michael E. DeBakey Stroke Program, Michael E. DeBakey VA Medical Center, Houston, TX
| |
Collapse
|
96
|
Lioutas VA, Alfaro-Martinez F, Bedoya F, Chung CC, Pimentel DA, Novak V. Intranasal Insulin and Insulin-Like Growth Factor 1 as Neuroprotectants in Acute Ischemic Stroke. Transl Stroke Res 2015; 6:264-75. [PMID: 26040423 DOI: 10.1007/s12975-015-0409-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/16/2015] [Accepted: 05/13/2015] [Indexed: 12/22/2022]
Abstract
Treatment options for stroke remain limited. Neuroprotective therapies, in particular, have invariably failed to yield the expected benefit in stroke patients, despite robust theoretical and mechanistic background and promising animal data. Insulin and insulin-like growth factor 1 (IGF-1) play a pivotal role in critical brain functions, such as energy homeostasis, neuronal growth, and differentiation. They may exhibit neuroprotective properties in acute ischemic stroke based upon their vasodilatory, anti-inflammatory and antithrombotic effects, as well as improvements of functional connectivity, neuronal metabolism, neurotransmitter regulation, and remyelination. Intranasally administered insulin has demonstrated a benefit for prevention of cognitive decline in older people, and IGF-1 has shown potential benefit to improve functional outcomes in animal models of acute ischemic stroke. The intranasal route presents a feasible, tolerable, safe, and particularly effective administration route, bypassing the blood-brain barrier and maximizing distribution to the central nervous system (CNS), without the disadvantages of systemic side effects and first-pass metabolism. This review summarizes the neuroprotective potential of intranasally administered insulin and IGF-1 in stroke patients. We present the theoretical background and pathophysiologic mechanisms, animal and human studies of intranasal insulin and IGF-1, and the safety and feasibility of intranasal route for medication administration to the CNS.
Collapse
Affiliation(s)
- Vasileios-Arsenios Lioutas
- Department of Neurology, Division of Cerebrovascular Diseases, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Palmer 127, Boston, MA, 02215, USA,
| | | | | | | | | | | |
Collapse
|
97
|
Parody E, Pedraza S, García-Gil MM, Crespo C, Serena J, Dávalos A. Cost-Utility Analysis of Magnetic Resonance Imaging Management of Patients with Acute Ischemic Stroke in a Spanish Hospital. Neurol Ther 2015; 4:25-37. [PMID: 26847673 PMCID: PMC4470974 DOI: 10.1007/s40120-015-0029-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Stroke has a high rate of long-term disability and mortality and therefore has a significant economic impact. The objective of this study was to determine from a social perspective, the cost-utility of magnetic resonance imaging (MRI) compared to computed tomography (CT) as the first imaging test in acute ischemic stroke (AIS). METHODS A cost-utility analysis of MRI compared to CT as the first imaging test in AIS was performed. Economic evaluation data were obtained from a prospective study of patients with AIS ≤12 h from onset in one Spanish hospital. The measure of effectiveness was quality-adjusted life-years (QALYs) calculated from utilities of the modified Rankin Scale. Both hospital and post-discharge expenses were included in the costs. The incremental cost-effectiveness ratio (ICER) was calculated and sensitivity analysis was carried out. The costs were expressed in Euros at the 2004 exchange rate. RESULTS A total of 130 patients were analyzed. The first imaging test was CT in 87 patients and MRI in 43 patients. Baseline variables were similar in the two groups. The mean direct cost was €5830.63 for the CT group and €5692.95 for the MRI group (P = not significant). The ICER was €11,868.97/QALY. The results were sensitive when the indirect costs were included in the analysis. CONCLUSION Total direct costs and QALYs were lower in the MRI group; however, this difference was not statistically significant. MRI was shown to be a cost-effective strategy for the first imaging test in AIS in 22% of the iterations according to the efficiency threshold in Spain.
Collapse
Affiliation(s)
| | - Salvador Pedraza
- Department of Radiology-IDI, IDIBGI, Hospital Doctor Josep Trueta, UDG, Girona, Spain
| | - María M García-Gil
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Catalunya, Spain
| | - Carlos Crespo
- Health Economics and Pricing, Boehringer Ingelheim, Sant Cugat del Valles (Barcelona), Spain
| | - Joaquín Serena
- Department of Neurology, Hospital Doctor Josep Trueta, Girona, Spain
| | - Antoni Dávalos
- Department of Neurology, Germans Trias i Pujol Hospital, Badalona, Spain
| |
Collapse
|
98
|
Mucke J, Möhlenbruch M, Kickingereder P, Kieslich PJ, Bäumer P, Gumbinger C, Purrucker J, Mundiyanapurath S, Schlemmer HP, Bendszus M, Radbruch A. Asymmetry of deep medullary veins on susceptibility weighted MRI in patients with acute MCA stroke is associated with poor outcome. PLoS One 2015; 10:e0120801. [PMID: 25849958 PMCID: PMC4388537 DOI: 10.1371/journal.pone.0120801] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 02/06/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Due to its sensitivity to deoxyhemoglobin, susceptibility weighted imaging (SWI) enables the visualization of deep medullary veins (DMV) in patients with acute stroke, which are difficult to depict under physiological circumstances. This study assesses the asymmetric appearance of prominent DMV as an independent predictor for stroke severity and outcome. MATERIALS AND METHODS SWI of 86 patients with acute middle cerebral artery (MCA) stroke were included. A scoring system from 0 (no visible DMV) to 3 (very prominent DMV) was applied for both hemispheres separately. A difference of scores between ipsi- and contralateral side was defined as asymmetric (AMV+). Occurrence of AMV+ was correlated with the National Institute of Health Stroke Scale (NIHSS) Score on admission and discharge, as well as the modified Rankin Scale (mRS) at discharge. Ordinal regression analysis was used to evaluate NIHSS and mRS as predictors of stroke severity, clinical course of disease and outcome. RESULTS 55 patients displayed AMV+ while 31 did not show an asymmetry (AMV-). Median NIHSS on admission was 17 (11-21) in the AMV+ group and 9 (5-15) in the AMV- group (p = 0.001). On discharge median NIHSS was 11 (5-20) for AMV+ and 5 (2-14) for AMV- (p = 0.005). The median mRS at discharge was 4 (3-5) in the AMV+ group and 3 (1-4) in AMV- (p = 0.001). Odds ratio was 3.19 (95% CI: 1.24-8.21) for AMV+ to achieve a higher mRS than AMV- (p = 0.016). CONCLUSION The asymmetric appearance of DMV on SWI is a fast and easily evaluable parameter for the prediction of stroke severity and can be used as an additional imaging parameter in patients with acute MCA stroke.
Collapse
Affiliation(s)
- Johanna Mucke
- University of Heidelberg, Department of Neuroradiology, INF 400, 69120 Heidelberg, Germany
| | - Markus Möhlenbruch
- University of Heidelberg, Department of Neuroradiology, INF 400, 69120 Heidelberg, Germany
| | - Philipp Kickingereder
- University of Heidelberg, Department of Neuroradiology, INF 400, 69120 Heidelberg, Germany
| | - Pascal J. Kieslich
- University of Mannheim, Department of Psychology, Schloss Ehrenhof Ost, 68131 Mannheim, Germany
| | - Philipp Bäumer
- University of Heidelberg, Department of Neuroradiology, INF 400, 69120 Heidelberg, Germany
| | - Christoph Gumbinger
- University of Heidelberg, Department of Neurology, INF 400, 69120 Heidelberg, Germany
| | - Jan Purrucker
- University of Heidelberg, Department of Neurology, INF 400, 69120 Heidelberg, Germany
| | - Sibu Mundiyanapurath
- University of Heidelberg, Department of Neurology, INF 400, 69120 Heidelberg, Germany
| | - Heinz-Peter Schlemmer
- German Cancer Research Center (DKFZ), Department of Radiology, INF 280, 69120 Heidelberg, Germany
| | - Martin Bendszus
- University of Heidelberg, Department of Neuroradiology, INF 400, 69120 Heidelberg, Germany
| | - Alexander Radbruch
- University of Heidelberg, Department of Neuroradiology, INF 400, 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), Department of Radiology, INF 280, 69120 Heidelberg, Germany
| |
Collapse
|
99
|
Lapchak PA. A cost-effective rabbit embolic stroke bioassay: insight into the development of acute ischemic stroke therapy. Transl Stroke Res 2015; 6:99-103. [PMID: 25637174 PMCID: PMC4359071 DOI: 10.1007/s12975-015-0386-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 01/06/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Paul A Lapchak
- Departments of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion Suite 8305, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA,
| |
Collapse
|
100
|
Li H, Liu X, Zhu Y, Liu Y, Wang Y. Magnolol derivative 002C-3 protects brain against ischemia-reperfusion injury via inhibiting apoptosis and autophagy. Neurosci Lett 2015; 588:178-83. [PMID: 25575794 DOI: 10.1016/j.neulet.2015.01.007] [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: 11/02/2014] [Revised: 01/02/2015] [Accepted: 01/05/2015] [Indexed: 12/28/2022]
Abstract
Neuroprotective agents can rescue ischemic penumbra in cerebral ischemia. However, the clinically effective neuroprotective agents for cerebral ischemic injury remain deficient in clinic so far. This study was undertaken to investigate the brain protective effect of 002C-3 and its potential mechanisms in rats, and its preliminary toxicity in mice. A transient middle cerebral artery occlusion (tMCAO) model in rats was used to evaluate its effect and mechanism, a dose limited experiment was used to evaluate its preliminary toxicity. 10-50μg/kg of 002C-3 (single iv bolus after reperfusion) significantly reduced neurological scores, infarct volumes and brain water contents, and the effect was more potent than that of magnolol under the same mole dose; 50μg/kg of 002C-3 significantly decreased the number of TUNEL-positive cells, reduced the activity of caspase-3, and lowered the autophagy-related proteins LC3-II and Beclin-1 level in I-R cerebral tissue. At 1000 times' dose of high effective dose (ip) 002C-3 failed to show evident toxicity in mice, and the mean body weight of mice treated with 002C-3 was almost the same as that of the vehicle control, but magnolol caused evident toxicity and death. In conclusion, 002C-3 has significant protective effect against cerebral ischemia-reperfusion injury; the effect is more potent than magnolol; this effect is maybe associated with its inhibition of both apoptosis and autophagy; its toxicity is greatly reduced compared to magnolol. These results provided data for its further research and development.
Collapse
Affiliation(s)
- Hongfei Li
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoyan Liu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuanjun Zhu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ye Liu
- Beijing Honghui New Medical Technology Co., Ltd. Beijing Daxing Biological Medicine Industry Base, Beijing 102600, China
| | - Yinye Wang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|