Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Stroke is the leading cause of disability and thesecond leading cause of death worldwide. With the global population aged 65 and over growing faster than all other age groups, the incidence of stroke is also increasing. In addition, there is a shift in the overall stroke burden towards younger age groups, particularly in low and middle-income countries. Stroke in most cases is caused due to an abrupt blockage of an artery (ischemic stroke), but in some instances stroke may be caused due to bleeding into brain tissue when a blood vessel ruptures (hemorrhagic stroke). Although treatment options for stroke are still limited, with the advancement in recanalization therapy using both pharmacological and mechanical thrombolysis some progress has been made in helping patients recover from ischemic stroke. However, there is still a substantial need for the development of therapeutic agents for neuroprotection in acute ischemic stroke to protect the brain from damage prior to and during recanalization, extend the therapeutic time window for intervention and further improve functional outcome. The current review has assessed the past challenges in developing neuroprotective strategies, evaluated the recent advances in clinical trials, discussed the recent initiative by the National Institute of Neurological Disorders and Stroke in USA for the search of novel neuroprotectants (Stroke Preclinical Assessment Network, SPAN) and identified emerging neuroprotectants being currently evaluated in preclinical studies. The underlying molecular mechanism of each of the neuroprotective strategies have also been summarized, which could assist in the development of future strategies for combinational therapy in stroke treatment.

Free full text 


Logo of nihpaAbout Author manuscriptsSubmit a manuscriptHHS Public Access; Author Manuscript; Accepted for publication in peer reviewed journal;
Exp Neurol. Author manuscript; available in PMC 2022 Jan 1.
Published in final edited form as:
PMCID: PMC7869696
NIHMSID: NIHMS1642861
PMID: 33144066

Emerging Neuroprotective Strategies for the Treatment of Ischemic Stroke: An Overview of Clinical and Preclinical Studies

Abstract

Stroke is the leading cause of disability and the second leading cause of death worldwide. With the global population aged 65 and over growing faster than all other age groups, the incidence of stroke is also increasing. In addition, there is a shift in the overall stroke burden towards younger age groups, particularly in low and middle-income countries. Stroke in most cases is caused due to an abrupt blockage of an artery (ischemic stroke), but in some instances stroke may be caused due to bleeding into brain tissue when a blood vessel ruptures (hemorrhagic stroke). Although treatment options for stroke are still limited, with the advancement in recanalization therapy using both pharmacological and mechanical thrombolysis some progress has been made in helping patients recover from ischemic stroke. However, there is still a substantial need for the development of therapeutic agents for neuroprotection in acute ischemic stroke to protect the brain from damage prior to and during recanalization, extend the therapeutic time window for intervention and further improve functional outcome. The current review has assessed the past challenges in developing neuroprotective strategies, evaluated the recent advances in clinical trials, discussed the recent initiative by the National Institute of Neurological Disorders and Stroke in USA for the search of novel neuroprotectants (Stroke Preclinical Assessment Network, SPAN) and identified emerging neuroprotectants being currently evaluated in preclinical studies. The underlying molecular mechanism of each of the neuroprotective strategies have also been summarized, which could assist in the development of future strategies for combinational therapy in stroke treatment.

Keywords: blood-brain barrier dysfunction, clinical trials, comorbidities, excitotoxicity, ischemic stroke, neuroinflammation, neuroprotection, neuroprotective agents, oxidative stress, preclinical studies

1. INTRODUCTION

Cerebrovascular accident or stroke is the second largest cause of death worldwide with an annual mortality rate of about 5.5 million. It is also the leading cause of disability worldwide with 50% of survivors being chronically disabled (Donkor, 2018; Katan and Luft, 2018). New research indicates that the occurrence of stroke is on the increase with one in four people worldwide to experience stroke in their lifetime (Collaborators et al., 2018). An aging population coupled with the burden of accumulating risk factors contributes to such increase in lifetime risk of stroke. In addition, increasing socioeconomic status in the developing countries has led to an epidemic rise in the risk factors for stroke in younger adults (Katan and Luft, 2018). These risk factors include atrial fibrillation, hypertension, hyperlipidemia, hyperhomocysteinemia, diabetes, smoking, lack of physical activity, unhealthy diet, abdominal obesity and alcohol consumption (Boehme et al., 2017; O’Donnell et al., 2016). However, hypertension is by far the most potent risk factor for stroke that accounts for ~54% of all strokes (Benjamin et al., 2019; Boehme et al., 2017; Lawes et al., 2008). Prevalence of hypertension in American adults is estimated to be ~45.4%, with an overall frequency of ~71.8% at age 60 years and older (Benjamin et al., 2019; Dorans et al., 2018). The absolute burden of hypertension in American adults has increased from ~86.6 million in 1999 to ~108 million in 2015 (Dorans et al., 2018). Notably, the global burden of hypertension has increased from 442 million in 1990 to 874 million in 2015, a ~97% increase, and it has been predicted that over the next two decades the incidence of stroke will increase by another ~24.9% (Forouzanfar et al., 2017; Heidenreich et al., 2011; Huffman and Lloyd-Jones, 2017; Kearney et al., 2005). Stroke is primarily classified as ischemic or hemorrhagic in nature. Ischemic stroke is caused by reduction of blood supply to a certain region of the brain due to obstruction of a blood veesel. In contrast, hemorrhagic stroke occurs due to the rupture of a blood vessel in the brain causing bleeding in the brain or the subarachnoid space (Amarenco et al., 2009; Donnan et al., 2008). Several studies have established that ischemic stroke is the most prevalent form of stroke worldwide (Bamford et al., 1991; Donkor, 2018; O’Donnell et al., 2010; Sarfo et al., 2018).

2. PATHOPHYSIOLOGY OF ISCHEMIC STROKE

The progression of ischemic brain damage following impaired blood flow involves the initial development of a core of irreversibly injured necrotic tissue within the affected vascular bed, followed by late phase injury development in the peri-infarct area, a potentially salvageable area surrounding the core (Astrup et al., 1981; Dirnagl et al., 1999; Ramos-Cabrer et al., 2011). The pathophysiology of ischemic brain damage involves the activation of a series of detrimental signaling cascades. Deprivation of oxygen and glucose supply to the brain tissue leads to immediate failure of energy-dependent ion pumps and channels resulting in the release of potentially toxic concentrations of excitatory neurotransmitters and subsequent death of the vulnerable neurons. The excessive release of the excitatory neurotransmitter glutamate is undoubtedly a prime element initiating the progression of brain damage through activation of N-methyl-D-aspartate (NMDA), Voltage gated (L-type), and quisqualate (Q) post synaptic receptor complexes (Pulsinelli, 1992). The influx of intracellular calcium through these receptors leads to the activation of a series of intracellular signaling cascades (e.g, mitochondrial depolarization, activation of cytosolic phospholipase A2, catalysis of arachidonic acid by cyclooxygenase-2 and activation of NADPH oxidase) resulting in increased free radical generation (Sun et al., 2018). The oxidative and nitrosative stress caused by free radicals are further accelerated by restoration of oxygen supply following reperfusion, inducing lipid peroxidation, protein oxidation and DNA damage during ischemia and reperfusion (Nelson et al., 1992; Sun et al., 2018). Additionally, oxidative stress induced release of damage-associated molecular pattern (DAMP) molecules triggers an inflammatory response involving activation of brain-intrinsic microglia, increased blood-brain barrier (BBB) permeability and peripheral immune cell infiltration (Banjara and Ghosh, 2017; Gulke et al., 2018). Increasing evidences indicate that the post-ischemic inflammation accounts for the secondary progression of brain damage, and the severity of stroke outcome under comorbidities depends on the extent of this inflammatory response (Chamorro et al., 2012; Lakhan et al., 2009; Veltkamp and Gill, 2016; Wang et al., 2007). A simplified overview of the ischemic cascade has been presented in Figure 1.

An external file that holds a picture, illustration, etc.
Object name is nihms-1642861-f0001.jpg

Schematic representation of ischemia induced signaling cascade in the brain that leads to necrosis, neurotoxicity and inflammation, which contributes to progression of ischemic brain injury. Abbreviations: DAMPs: damage-associated molecular patterns; NMDAR: N-methyl-D-aspartate receptor; AMPAR: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; cPLA2: cytosolic phospholipase A2; COX2: cyclooxygenase-2; nNOS: neuronal nitric oxide synthase; ROS: reactive oxygen species; RNS: reactive nitrogen species; BBB: blood-brain barrier.

Despite advances in understanding the pathophysiology of stroke, successful treatment remains a major challenge in clinical medicine. Reperfusion with recombinant tissue plasminogen activator (rtPA) remains the only pharmacological therapy (Cohen et al., 2011; Del Zoppo et al., 2009; Kwiatkowski et al., 1999). In a small number of cases surgical recanalization is also possible (Choi et al., 2006; Cohen et al., 2011; Prabhakaran et al., 2015; Smith et al., 2008). However, rapid reperfusion, although necessary for restoration of brain metabolic activity, is also associated with additional risks (Bai and Lyden, 2015; Khatri et al., 2012; Mizuma et al., 2018). The development of neuroprotection strategies to protect brain cells from both ischemia and reperfusion injury as well as to amplify the time window for thrombolytic treatment is therefore an important goal (Chamorro et al., 2016).

3. PAST CHALLENGES IN DEVELOPING NEUROPROTECTIVE STRATEGIES

Neuroprotectants refers to any agent(s) capable of reducing ischemic brain injury by antagonizing detrimental molecular events in the brain rather than improving cerebral blood flow. Preclinical research for several decades has demonstrated the potential benefit of neuroprotection in experimental stroke models (O’Collins et al., 2006). However translation of such neuroprotective treatments from animal models to humans is yet to yield a positive result (Braeuninger and Kleinschnitz, 2009; Sacchetti, 2008).

Failure to include stroke models with significant comorbidities and sub-optimal design of many of the pre-clinical studies could have contributed to the high failure rate. In many of the earlier studies, neuroprotective agents were tested only in young animals, which do not mimic all aspects of stroke in older humans (Chen et al., 2010a; DiNapoli et al., 2008; Jin et al., 2004). In only 6% of human patients, stroke occurs in isolation (no comorbidities), and high prevalence of one or more pre-existing comorbidities is known to worsen stroke outcome in patients (Fischer et al., 2006; Gallacher et al., 2014; Guthrie et al., 2012; Karatepe et al., 2008). Moreover, stroke outcome measures in many of these earlier studies have been limited to final infarct size measurement at 24h and relatively limited neurological testing over a short-time period. However, the extent of lesion and functional deficits may vary over prolonged time periods (Colbourne et al., 1999; Virley et al., 2000). As a result, Stroke Therapy Academic Industry Roundtable (STAIR) recommendations now emphasizes a focus on agents with multiple mechanism(s) of action in neuroprotection, assessment of efficacy when treatment is combined with thrombolytic agents, assessment of long-term recovery using non-invasive magnetic resonance imaging and complex sensorimotor and cognitive tasks, as well as inclusion of experimental models of stroke with comorbidities and advanced age in both sexes (Albers et al., 2011; Fisher et al., 2005). From the translational standpoint, it is also important to consider the need for multicenter randomized pre-clinical trials in animal models of stroke (Balduini et al., 2016; Dirnagl and Fisher, 2012; Kellner et al., 2016), similar to what has been adapted in human clinical trials. Preclinical studies that meet these stringent criteria should also be presented in the public domain before moving to clinical trials.

The abundance of failed clinical trials also points toward several caveats/deficiencies in the quality or scope of these trials (Ginsberg, 2008; Xiong et al., 2018). The most important among these caveats is the time of administration of neuroprotective agents. For most of these trials patients were enrolled beyond 4h after stroke onset, which is beyond the optimal time window (4–6h) for efficacious neuroprotection (Saver, 2013). Pre-hospital administration of neuroprotectants in the field by paramedics may allow the therapy to be started at the earliest opportunity and slow down the progression of ischemic brain injury. Low rates of recanalization may have also contributed to the lack of efficacy of neuroprotective agents in earlier clinical trials. In the current thrombectomy era, cotreatment with neuroprotective agents during recanalization could augment the delivery of neuroprotective agents to the targeted brain regions and further enhance the efficacy of stroke therapy. Furthermore, it needs to be considered that all stroke patients may not benefit from a given neuroprotective agent. Also, additional studies are needed to identify serological biomarkers for different subgroups of stroke patients. This information could then be used to determine the efficacy of potential neuroprotective agents in animal models of stroke. Optimal patient selection based on such biomarkers could decrease the likelihood of failure in clinical trials. Thus, a renewed approach towards development of novel neuroprotectants must consider the lessons learned from all the earlier studies, both pre-clinical and clinical, to devise a plausible path to success.

In this review, we have compiled the list of (1) recently completed clinical trials with neuroprotectants performed between 2015 through 2020, (2) ongoing clinical trials, (3) NIH-initiated search for neuroprotectants as part of the Stroke Preclinical Assessment Network (SPAN) and (4) emerging neuroprotectants. We have further discussed the mechanism(s) of action and therapeutic approaches for these potential neuroprotectants.

4. RECENTLY COMPLETED CLINICAL TRIALS FOR STROKE NEUROPROTECTION

Several clinical trials were initiated in the last five years to test the beneficial effects of multiple neuroprotective agents. For most of these agents, some experimental data were available on the mechanism(s) of action, functional outcome following treatment in rodent models of stroke, efficacy in stroke models with comorbidities and use at longer intervals after ischemic injury. As inflammatory mediators play a crucial role in the progression of ischemic brain damage, an important target for these potential neuroprotective agents has been the post-stroke inflammatory pathway, although some of them have diverse effects. In addition, two of these neuroprotective agents targets excitotoxicity and oxidative stress signaling pathway (Table 1).

Table 1:

Recently completed (last 5 yeas) clinical trials to assess neuroprotection in ischemic stroke.

AgentTrial NameTrial No./Phase/LocationTime of intervention (post-stroke)Proposed mechanisms of neuroprotection
Human urinary kallidinogenase (HUK)-NCT03431909 Phase IV ChinaWithin 72hAnti-oxidative and anti-inflammatory agent

StatinsASSORTNCT02549846 Phase IV JapanWithin 24h of admission; Atorvastatin (20mg), or Pitavastatin (4mg) or Rosuvastatin (5mg)Pleiotropic effect with anti-inflammatory and anti-oxidant activity; reduces BBB dysfunction and enhances cerebral blood flow
NeuSTART2NCT01976936 Phase II USAWithin 0h to 24h of symptom onset; Lovastatin - 640mg/day for 3 days

Edaravone-NCT02430350 Phase III China/JapanEvery 12h for 2 weeks (37.5mg / dose)Potent anti-oxidant with other neuroprotective properties

NA-1 (nerinetide)ESCAPENCT02930018 Phase III Canada & USAAs soon as they meet the enrollment criteria, within 12h (2.6 mg/kg)Targets NMDA receptor mediated excitotoxicity

3K3A-APCRHAPSODYNCT02222714 Phase II USAWithin 30min to 120min of tPA infusion or mechanical thrombectomy; q12h,5 doses in 3 days; (120μg, 240μg, 360μg or 540μg/kg)Anti-inflammatory agent that also suppresses intracerebral hemorrhage

GM602GMAISNCT01221246 Phase II USAWithin 18h; three consecutive daily doses (320mg or 480mg)Multi-target drug with anti-inflammatory, antioxidative and antiapoptotic properties

NatalizumabACTION2NCT02730455 Phase II Europe & USAWithin 9h or 9-24h Dose - 300mg or 600mg IVAnti-inflammatory agent

Vinpocetine-NCT02878772 Phase II ChinaBetween 4.5h to 48h after symptom onset and 1h (30mg)Cerebral vasodilator with anti-oxidative and anti-inflammatory properties

Human urinary kallidinogenase (HUK) is a tissue kallikrein extracted from urine (Savica et al., 2011; Yousef and Diamandis, 2001). It is a glycoprotein that can cleave the low molecular weight protein kininogen to produce the potent vasodilator peptide kinin, which can trigger a series of biological effects (Chao and Chao, 2005). Accumulating evidence indicates that tissue kallikrein is a promising target for the treatment of acute ischemic stroke. Studies in animal models of acute ischemic stroke showed that HUK reduces infarct volume and improves neurological deficits (Chao and Chao, 2006; Wei et al., 2018). The underlying mechanisms involve increased local vasodilation and neuroprotection (Chen et al., 2010b; Emanuelia and Madeddu, 2003; Han et al., 2015; Yang et al., 2017c). HUK confers neuroprotection as an anti-oxidative and anti-inflammatory agent, through suppression of toll-like receptor 4 and the nuclear factor-kappa B (TLR4/NF-κB) signaling pathway that induces the expression of pro-inflammatory genes. It also increases the expression of transforming growth factor beta 1 (TGF-β1), which is known to attenuate neuroinflammation (Cekanaviciute et al., 2014; Dong et al., 2016; Yang et al., 2017c). Additional studies in ex vivo models of hypoxic injury have further demonstrated that HUK could inhibit glutamate-induced oxidative injury and neurotoxicity (Liu et al., 2011; Liu et al., 2009). A meta-analysis of 24 small scale randomized controlled trials in acute ischemic stroke with HUK treatment have reported improved neurological function and long-term outcome. However, treatment related adverse events were also observed in 15 trials with transient hypotension being the most common one (Zhang et al., 2012a). For a comprehensive re-evaluation of the safety and efficacy of HUK, a large scale open-label, singlearm, multicenter phase IV trial (NCT02562183) was subsequently designed (Ni et al., 2017). A more recent analysis of pooled data from randomized double-blind placebo-controlled phase IIb and phase III clinical trial have showed that HUK is safe and provides potential clinical benefits as a treatment of acute ischemic stroke regardless of age, sex and comorbidities when treatment is initiated within 48h of symptom onset (Dong et al., 2020b). In addition, a phase IV clinical trial (NCT03431909) to assess the efficacy of HUK in acute ischemic stroke has also been recently completed, but outcome of this trial has still not been reported. HUK has been approved by China‟s State Food and Drug Administration to be used for treatment of mild to moderate stroke (Wu et al., 2017a).

Statins are a group of drugs that inhibit 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase, an enzyme involved in cholesterol synthesis (Vaughan et al., 2000). Statins are either fungal-derived (lovastatin, pravastatin and simvastatin) or fully synthetic compounds (atorvastatin, cerivastatin, pitavastatin, rosuvastatin and fluvastatin) (Schachter, 2005). Although the different statins vary in their efficacy for loweing cholesterol, currently they are the most effective agents available for treating hyperlipidemia. They are also effective in decreasing cardiovascular-related morbidity and mortality in individuals with or without coronary artery disease (Lee et al., 2019; Zhou and Liao, 2009). More recently, studies in experimental animal models of stroke have shown that statins can also reduce ischemic infarct size when administered before or after stroke as well as with or without thrombolytic agents (Amin-Hanjani et al., 2001; Balduini et al., 2001; Elkind, 2006; Endres et al., 1998; Gertz et al., 2003; Kawashima et al., 2003; Laufs et al., 2002; Sironi et al., 2003). The efficacy of statins in reducing ischemic brain damage have been shown to be independent of its cholesterollowering ability. In these studies, statins upregulated endothelial nitric oxide synthase (eNOS) in the cerebral vasculature, particularly in the ischemic penumbra, thereby increasing cerebral blood flow and limiting neuronal loss. Treatment with statins also provided protection against post-stroke BBB dysfunction by attenuating the expression of adhesion molecules (Mayanagi et al., 2008; Pruefer et al., 1999; Romano et al., 2000b). In addition, statins have also been shown to confer neuroprotection in the rodent ischemic brain through reduced generation of superoxide radicals and inhibition of NFκB-mediated expression of inflammatory cytokines (Cui et al., 2009; Hong et al., 2006; Kawai et al., 2011; Romano et al., 2000a; Sironi et al., 2006; Weitz-Schmidt, 2002). These pleiotropic effects of statins also account for the reduced infarct size observed in rodent models of ischemic stroke with hypertension (Garcia-Bonilla et al., 2012; Nagotani et al., 2005). Recently, clinical trials have been initiated to evaluate the therapeutic potential of statins in ischemic stroke patients. Phase I clinical trial (NeuSTART) first established the efficacy and feasibility of using lovastatin in stroke patients (Elkind et al., 2008; Elkind et al., 2009). A phase II randomized, blinded and controlled study (NeuSTART2) that was recently completed, assessed the efficacy of short-term administration of a high dose of levostatin (640 mg/day) for 3 days after stroke when compared with patients who were treated with the standard dose of levostatin (80 mg/day) before and after stroke. Functional outcome, inflammatory markers and lipid levels were also assessed 3-months post stroke. However, the outcome of this clinical trial has not yet been published. Another multicenter randomized controlled trial (ASSORT) also completed recently showed that delayed statin therapy (7 days following stroke) was as efficient as statin treatment administered during early acute phase of ischemic stroke (within 24h) in its ability to alleviate physical disability assessed 3-months after stroke onset (Yoshimura et al., 2017).

Edaravone, also known as MCI-186 (3-methyl-1-phenyl-2-pyrazolin-5-one) is a low molecular weight free radical scavenger that can readily cross the blood-brain barrier (Yoshida et al., 2006). Numerous studies in experimental rodent models of ischemic stroke have demonstrated that pre-treatment or post-treatment with edaravone is effective in decreasing infarct volume and improve neurological function (Kitagawa, 2006; Nishi et al., 1989; Shichinohe et al., 2004; Watanabe et al., 1994; Wu et al., 2000). Edaravone exerts its neuroprotective effects primarily through its anti-oxidant activity (Banno et al., 2005; Higashi, 2009; Lee et al., 2010; Yamamoto et al., 1997). However, the therapeutic potential of edaravone in the treatment of stroke has also been attributed to its ability to regulate additional deleterious cascades, which includes inhibition of lipid oxidation, endothelial expression of nitric oxide synthase by oxidized LDL, inflammatory response caused by the release of high-mobility group box-1 protein from neurons, microglia-induced neurotoxicity and matrix metalloproteinase-9 (MMP-9) activation (Banno et al., 2005; Higashi et al., 2006; Yagi et al., 2009; Yoshida et al., 2005; Zhang et al., 2005). It has also been suggested that edaravone may prevent post-stroke edema by reducing the levels of endothelial growth factor and aquaporin-4 in astrocytes (Kikuchi et al., 2009). Although these findings suggest that edaravone is a multi-target neuroprotective agent that could be beneficial for stroke treatment, information regarding its efficacy in preclinical models with comorbidities and long-term outcome have not been reported. In clinical studies, safety and pharmacokinetic profile evaluation of edaravone have established that it is well tolerated in normal volunteers following administration of a single or multiple doses (Shibata et al., 1998). Analysis of multiple clinical trials evaluating the efficacy of edaravone on embolic, thrombotic and lacunar strokes have also established beneficial effects of treatment with regards to functional outcome and survival parameters, which has been extensively discussed in two earlier review articles (Lapchak, 2010; Rolain et al., 2019). These studies showed that edaravone is most effective when administered within 24h following a stroke, and extended treatment is required for maximum efficacy. However, several adverse side effects of edaravone have also been reported in stroke clinical trials, of which increased renal toxicity is of major concern, although it seems to be reversible in some patients (Edaravone Acute Infarction Study, 2003; Hishida, 2007, 2009; Shinohara et al., 2009). A few clinical studies documented the use of edaravone in combination with thrombolytics and the most intriguing finding from one such study is that treatment with edaravone prior to intravenous administration of thrombolytics reduced intracerebral hemorrhage (Enomoto et al., 2019). Based on these findings edaravone has been approved for the treatment of acute ischemic stroke in Japan, but it is still not considered for standard treatment globally (Kikuchi et al., 2010; Kikuchi et al., 2017; Lapchak, 2010). To further assess its efficacy as a neuroprotective agent, a large-scale clinical trial (NCT02430350) was recently completed to compare the effects of treatment with edaravone alone or in combination with borneol, an anti-inflammatory agent, for which the outcome has still not been reported.

NA-1 (nerinetide; TATNR2B9c) is a cell-permeable peptide comprised of nine COOH-terminal residues of NR2B subunit of NMDA receptor. It interacts with and disrupts the binding of synaptic protein PSD-95 to the NR2B subunit of NMDA receptor and to the PDZ domain of neuronal nitric oxide synthase (nNOS). Disruption of these protein-protein interaction by NA-1 attenuates the neurotoxic signaling cascades downstream of NMDAR activation that includes excessive neuronal calcium ion influx and excitotoxic nitric oxide production (Aarts et al., 2002; Ballarin and Tymianski, 2018; Sattler et al., 1999; Soriano et al., 2008). Thus, NA-1 prevents excitotoxic cell death and subsequent brain damage without blocking normal synaptic function of NMDA receptors. Long-term evaluation of stroke outcomes in rodent and non-human primate models of focal ischemia have further showed that treatment with NA-1, even when administered after the onset of stroke, can reduce infarct size along with improvement in neurological deficits (Bratane et al., 2011; Cook et al., 2012; Sun et al., 2008). The efficacy and safety of NA-1 was also recently evaluated in a multi-centre, double blind randomized controlled clinical trial (ESCAPE). The study enrolled stroke patients with large vessel occlusion within a 12h treatment window in 48 acute care hospitals in eight countries. The findings show that treatment with a single dose of NA-1 along with endovascular thrombectomy failed to improve long-term stroke outcome when compared with patients receiving pacebo. However, in a sub-population of patients who received NA-1 treatment without any thrombolytics, a lower risk of mortality, significant reduction in infarct volume and improved functional outcome was observed (Hill et al., 2020). The authors hypothesized that in patients receiving both the thrombolytic agent (tPA) and NA-1, it is possible that alteplase was activating proteases that were digesting the drug. As such, further studies with this promising neuroprotectant are necessary to extensively explore its neuroprotective effects.

3K3A-activated protein C (APC) is an analogue of the endogenous blood serine protease APC where 3 lysine residues (residues 191–193) were replaced by 3 alanine residues (Zlokovic and Griffin, 2011). APC is derived from the protein C zymogen produced by the liver and it has both anticoagulant and cytoprotective properties (Rezaie, 2010). Earlier studies have established that the vasculoprotective, neuroprotective and anti-inflammatory activities of APC protects neurons and cerebrovascular endothelium from stroke injury (Guo et al., 2009; Shibata et al., 2001; Wang et al., 2009; Zlokovic et al., 2005). However, its anticoagulant activity increases the risk of intracerebral hemorrhage and is a significant side-effect in the clinical application of APC (Nadel et al., 2007). To minimize the risk of bleeding, 3K3A-APC was designed that lacks >90% of the anti-coagulant activity, while maintaining the cytoprotective activies of APC (Guo et al., 2009; Mosnier et al., 2004; Wang et al., 2009; Williams et al., 2012). In animal models of ischemic stroke, treatment with 3K3A-APC has been shown to reduce infarct size and improve functional outcome, even when administered several hours after the onset of the insult (Ellery et al., 2019; Wang et al., 2012). When 3K3A-APC was administered along with tPA, it also eliminated tPA-induced intracerebral hemorrhages and extended the therapeutic time window of treatment with tPA (Wang et al., 2012). In addition, 3K3A-APC has also been shown to be effective in the treatment of stroke in both aged and hypertensive animals (Wang et al., 2013). The mechanisms of 3K3A-APC-mediated neuroprotective and anti-inflammatory activities have been partly elucidated (Griffin et al., 2018). Studies in neuronal cultures have highlighted the requirement of protease activated receptors 1 and 3 (PAR1 and PAR3) in 3K3A-APC mediated attenuation of NMDAR mediated neurotoxicity (Guo et al., 2004). In vivo studies have further showed that APC mediated reduction in ischemic infarct volume and intracerebral hemorrhage during combination therapy with tPA requires activation of PAR1-mediated intracellular signaling (Cheng et al., 2006; Liu et al., 2004; Zlokovic and Griffin, 2011). In addition, suppression of NLRP3 inflammasome signaling is also a key contributor to 3K3A-APC‟s anti-inflammatory action (Guo et al., 2016; Guo et al., 2018; Nazir et al., 2017). Based on its strong neuroprotective and anti-inflammatory activities, preclinical safety and pharmacokinetic studies with 3K3A-APC were carried out in both mice and monkeys to ensure the feasibility of 3K3A-APC for stroke therapy in patients (Williams et al., 2012). A phase I clinical trial in healthy volunteers have further established that the drug is safe up to a maximum dose of 0.54mg/kg without any clinical manifestations of bleeding or bruising (Lyden et al., 2013). A recently completed Phase II clinical trial (RHAPSODY) in moderate to severe ischemic stroke patients, where 3K3A-APC treatment was initiated following tPA infusion or mechanical thrombectomy, also showed that the drug is well tolerated in stroke patients and there is a trend toward lower hemorrhage rate when compared to placebo-treated patients (Lyden et al., 2019). These promising findings have laid the groundwork for additional clinical trials in a larger cohort of stroke patients in the future.

GM6, developed by Genervon Biopharmaceuticals, is a peptide-based multi-target drug that is believed to reduce inflammation and limit neuronal degeneration (Swindell et al., 2018). It is a six amino acid peptide that represents the smallest active site of an endogenous neurotrophin, motoneurotrophic factor (MNTF) that is highly specific for the human nervous sytem and has maximum expression in the ninth week of human fetal development (Di and Huang, 1998; Valko et al., 2018; Yu et al., 2008). Some of its observed functions include motor neuron differentiation, maintenance and survival (Di and Huang, 1998; Di et al., 1997; Yu et al., 2008; Zhou et al., 1994). During injuries and disorders of the central and peripheral nervous sytems, the anti-apoptotic, antioxidative and anti-inflammatory functions of MNTF is thought to create a permissive environment for nerve regeneration and repair (Wang et al., 1995; Yu et al., 2008; Zhou et al., 1997; Zhou et al., 1993). More recent studies have shown that synthetic GM6 has similar activity and demonstrate similar neurotrophic activities as the parent molecule (Chau, 2005; Chau and Ko, 2007). It is anticipated that GM6 have a complex mechanism of action involving activation of multiple receptors and downstream signaling cascades (Yu et al., 2008). However, the exact mechanisms of action of GM6 is yet to be determined. GM6 has also been reported to reduce infarct volume and behavioral deficits in a ischemia/reperfusion injury mouse model (Yu et al., 2008). GM602, a FDA-approved proprietary form of GM6 was recently tested in a phase II clinical trial (GMAIS) to evaluate its efficacy and safety in treatment of ischemic stroke, the outcome of which is yet to be released.

Natalizumab is a humanized antibody against the cell adhesion glycoprotein alpha-4 integrin (CD49d) that is expressed on the surface of lymphocytes and monocytes and facilitates their adhesion to the endothelium (Lobb and Hemler, 1994; Rice et al., 2005). The cellular effect of natalizumab is to block the transmigration of lymphocytes and monocytes into the central nervous system (Rudick and Sandrock, 2004; Tubridy et al., 1999). The effectiveness of targeting alpha-4 integrin for stroke treatment in experimental ischemic stroke models have had mixed outcomes. Some studies have shown that blocking alpha-4 integrin reduces peripheral immune cell infiltration and subsequent reduction in infarct size under both normotensive and hypertensive conditions, while others have failed to show efficacy with treatment (Becker et al., 2001; Langhauser et al., 2014; Liesz et al., 2011b; Llovera et al., 2015; Relton et al., 2001). The results from a clinical trial (ACTION) on the safety and efficacy of a single dose of natalizumab (300 mg) given within 9h of acute ischemic stroke established the safety and feasibility of the study. However, with treatment at this dose the study outcome was negative for the efficacy endpoint (Elkins et al., 2017). Based on these findings, a second clinical trial (ACTION 2) was started in 2016 to evaluate the efficacy of a higher dose of natalizumab in stroke treatment. According to a report from Biogen, the trial failed to meet the efficay endpoint and the company decided not to pursue further development of natalizumab for stroke treatment.

Vinpocetine is a synthetic ethyl-ester derivative of the alkaloid apovincamine, which is isolated from Vinca minor leaves, commonly known as periwinkle plant (Al-Kuraishy et al., 2020). It was initially developed for the treatment of cerebrovascular diseases. A number of studies in rodent models of stroke have now established the protective effects of vinpocetine against brain damage caused by ischemia (Erdo et al., 1990; Jincai et al., 2014; Rischke and Krieglstein, 1991; Sauer et al., 1988; Wu et al., 2017b; Zhao et al., 2020). The neuroprotective function of vinpocetine has been attributed to multiple mechanisms of action (Zhang et al., 2018). In the brain, vinpocetine improves blood flow through its inhibition of phosphodiesterase 1 (PDE1) that catalyzes the degradation of cAMP and cGMP, two known vasodilators (Al-Kuraishy et al., 2020; Medina, 2011; Paterno et al., 1996; Wu et al., 2017b; Zhang et al., 2018). It also prevents neurotoxic calcium and sodium elevation by inhibiting neuronal voltage-gated sodium channel (Molnar and Erdo, 1995; Sitges et al., 2005; Sitges et al., 2011). In addition, vinpocetine has been shown to attenuate oxidative stress through inhibition of lipid peroxidation and free radical generation (Deshmukh et al., 2009; Santos et al., 2000; Zhao et al., 2020). More recent studies have further demonstrated that vinpocetine is a potent anti-inflammatory agent as an inhibitor of IκB kinase, which plays a critical role in NF-κB-dependent inflammatory response (Jeon et al., 2010; Wang et al., 2014; Wu et al., 2017b; Zhao et al., 2020). Vinpocetine can cross the BBB after oral or intravenous administration (Gulyas et al., 2002a; Gulyas et al., 2002b) and is widely used in many European and Asian countries for prevention of cerebrovascular disorders and cognitive impairments (Wu et al., 2017b; Zhang et al., 2018). It is also available as dietary supplements worldwide (Zhang et al., 2018). However, there is no evidence to support the routine use of vinpocetine in ischemic stroke patients. So far, the safety and feasibility of administering vinpocetine in ischemic stroke patients has been established in a previously conducted pilot study (Feigin et al., 2001). A meta-analysis reviewing the effects of vinpocetine treatment on acute ischemic stroke was inconclusive as most of the studies analyzed did not use double-blinded approaches and randomization. The two studies that used appropriate designs did not show significant differences between groups and were based on small samples (Bereczki and Fekete, 2008). As such, additional clinical trials are still needed to determine if vinpocetine treatment can reduce mortality and improve stroke outcome in surviving patients. A phase II multicenter randomized clinical trial (NCT02878772) in a small number of ischemic stroke patients was recently completed to evaluate the efficacy of vinpocetine on stroke outcome, where treatment was initiated between 4.5h to 48h after after the onset of symptoms. However, the findings of this trial has not been reported yet.

This comprehensive review of the neuroprotective agents assessesd in randomized clinical trials within the last five years highlights important differences in their mechanisms of action. While most of them targeted more than one pathophysiological cascade involved in stroke-induced brain damage, some of them have primarily focused on the inflammatory or excitotoxic pathways. Although the outcome is still unknown for many of these clinical trials, it has been reported that the anti-inflammatory agent, Natalizumab and the anti-excitotoxic agent, NA-1 have failed in phase II and phase III clinical trials, respectively. Natalizumab primarily reduces peripheral immune cell infiltration without any effect on inflammatory processes involving microglial activation. The failure of Natalizumab indicates that for anti-inflammatory therapies to be successful, it is important to develop strategies that could modulate the activity of both microglia and infiltrating immune cells. Since post-stroke inflammation can be both detrimental and beneficial for infarct resolution, a better understanding of the temporal and spatial dynamics of the pathogenic immune signaling involving both resident microglia and infiltrating immune cells is also necessary for the development of next-generation immune therapies. On the other hand, the failure of NA-1 suggests that drugs that will only prevent excitotoxic neuronal cell death without effecting the non-neuronal cells, which contributes substantially to the post-stroke secondary injury, will also not be successful. Collectively, these two failures highlights the importance of pleiotropic multi-target drugs for stoke treatment.

5. ONGOING CLINICAL TRIALS FOR STROKE NEUROPROTECTION

As our understanding of the mechanisms of acute ischemic stroke continues to advance, a variety of additional drugs with neuroprotective properties have been developed and are being evaluated in ongoing clinical trials (Table 2). Interestingly, most of these neuroprotective agents target multiple aspects of the neurodegenerative cascade and, therefore, might have synergistic effects against ischemic injury.

Table 2:

Ongoing clinical trials to assess neuroprotection in ischemic stroke.

AgentTrial NameTrial No./Phase/ LocationTime of intervention (post-stroke)Proposed Mechanisms
GinkgolideGIANTNCT03772847 China, Phase IVTo be administered along with tPAReduces BBB permeability, ER stress, inflammation and oxidative stress

Butylphthalide (NBP)-NCT03394950 Phase IV, ChinaWithin 4.5h; infusion for 14 days; 25mg/dayReduces oxidative damage, inflammation, apoptosis and mitochondrial dysfunction
EBCASNCT03539445 Phase III, ChinaWithin 6h: 100ml twice/day for 14 days; 60mg/day from day 15 to day 90
-NCT02905565 Phase II, USAWithin 12h; for 30 days; 800mg/day

BIIB093 (IV Glibenclamide)CHARMNCT02864953 USA, Phase IIIBolus within 10h followed by continuous intravenous infusion for 72h.Reduces edema and inflammation

HLCM051 (Multi Stem)MASTERS-2NCT03545607 USA, Phase IIIWithin 18-36h; 1.2 billion HLCM051 cellsReduces peripheral inflammatory response
TREASURENCT02961504 Japan, Phase II/IIIWithin 18-36h; 1.2 billion HLCM051 cells

Imatinib-NCT03639922 Phase III SwedenWithin 8h; for 6 days; 800mg/dayAmeliorates neuroinflammation by preserving BBB integrity.

Exenatide (GLP-1R agonist)TEXAISNCT03287076 Australia, Phase IIWithin 9h; 5 μg, twice daily for 5 daysReduces oxidative stress, inflammation and edema

JPI-289 (Amelparib)_NCT03062397 Phase II, KoreaWithin 24h (low or high dose)Attenuates NAD and ATP depletion, mitochondrial dysfunction and immune response

Neu2000SONICNCT02831088 Korea, Phase IIWithin 8h (750mg); nine follow-up infusion at 12h intervals (500mg)NR2B-selectve NMDA receptor antagonist and antioxidant

Remote ischemic conditioning (RIC)RIC-ACSNCT03868007 ChinaWithin 4h, twice daily for 14 daysTriggers anti-oxidative, anti-inflammatory, and mitochondria modulatory protective effects
RICAMISNCT03740971 ChinaWithin 48h, twice daily
RESISTNCT03481777 DenmarkAfter 6h, twice daily for 7 days
REMOTECATNCT03375762 SpainWithin 8h, prehospital setting - single treatment

Ginkgolides are a unique class of diterpenoids isolated from the roots and leaves of the ‘fossil’ tree Ginkgo biloba L (Jacobs and Browner, 2000). Ginkgolides A, B, C, J, K, L and M are the pharmacologically active diterpene lactones that have been well characterized (Stromgaard and Nakanishi, 2004; van Beek, 2005). They have many pharmacological activities that include selective and potent antagonism to platelet activating factor (PAF), as well as selective antagonism to the receptors for the neurotransmitters glycine and γ-aminobutyric acid (Ivic et al., 2003; Jensen et al., 2007; Koch, 2005; Zeng et al., 2013). Studies using animal models of both transient and permanent cerebral ischemia have shown that ginkgolides and their derivatives could attenuate neuronal injury and reduce brain infarct size (Lv et al., 2011; Ma et al., 2012; Nada and Shah, 2012; Shu et al., 2016; Sun et al., 2011b; Zhou et al., 2017). Additional studies have indicated that ginkgolides may also facilitate post-stroke recovery in hyperlipidemic rats, indicating its efficacy in experimental stroke models with comorbidities (Fang et al., 2015; Huang et al., 2012). The neuroprotective effects of ginkgolides on cerebral ischemia has been attributed to its antagonism to PAF, reduction in BBB permeability, inhibition of brain edema and inflammation, and scavenging oxygen free radicals (Feng et al., 2019). Clinical trials in stroke patients have also established the safety, feasibility and efficacy of ginkgolides in acute ischemic stroke (Dong et al., 2020a; Li et al., 2017b; Oskouei et al., 2013). Based on these findings, a large scale clinical trial (GIANT) in stroke patients is currently evaluating the efficacy of combined treatment with ginkgolide and intravenous thrombolysis. However, the mechanisms through which ginkgolides confers neuroprotection against ischemic brain injury are still in the early stages of investigation (Feng et al., 2019).

1–3-N-Butylphthalide (1-NBP), an active ingredient in the seeds of celery and its racemic form DL-3-N-butylphthalide (DL-NBP) have shown significant neuroprotective effects in stroke (Abdoulaye and Guo, 2016). NBP exerts its efficacy by targeting several pathways that include reduction in post-ischemic oxidative damage, neuronal apoptosis, mitochondrial dysfunction and inflammation (Chen et al., 2018; Hu et al., 2014; Lan et al., 2015; Li et al., 2018; Wang et al., 2018; Yan et al., 2017; Yang et al., 2019a). Additional studies have indicated that NBP may also facilitate post-stroke recovery by promoting neurogenesis (Sun et al., 2017). These studies have utilized both rodent models of permanent and transient focal cerebral ischemia to evaluate either short-term or long-term efficacy of NBP, and at least two such studies used hypertensive rats to evaluate the efficacy of post-stroke NBP treatment (Liao et al., 2009; Zhang et al., 2012b). Efficacy and safety of NBP have also been evaluated in multiple clinical trials in China and it is clinically-approved as an anti-ischemic drug in China (Abdoulaye and Guo, 2016; Cui et al., 2013; Xu et al., 2019; Zhang et al., 2020; Zhang et al., 2017). It is currently undergoing phase II clinical trial in USA (NCT03539445) as well as a phase III/IV clinical trials in China (NCT03394950) to evaluate its multitargeted effect in ischemic stroke.

BIIB093 is an intravenous (IV) formulation of glibenclamide (also known as glyburide). Glibenclamide is a sulfonylurea class of drugs that has been in clinical use as an oral hypoglycemic agent for the treatment of type 2 diabetes since 1969 (Feldman, 1985; Kurland et al., 2013; Marble, 1971) as it promotes the release of insulin by blocking pancreatic KATP [sulfonylurea receptor 1 (Sur1)-Kir6.2] channels (Kramer et al., 1995; Panten et al., 1996). Glibenclamide has received renewed attention in the last decade due to its neuroprotective effect in CNS injury, where its principal target is the transient receptor potential melastatin 4 (Trpm4), a non-slecetive cation channel that has been shown to be associated with Sur1 to form the Sur1-Trpm4 channels (Simard et al., 2006; Woo et al., 2013). Sur1-Trpm4 channels have been shown to be upregulated following ischemia in ischemic endothelial cells, neurons and glia, and has been associated with cytotoxic edema, BBB breakdown and formation of vasogenic edema (Mehta et al., 2015). Additional studies have also demonstrated that activated microglia in the ischemic lesion core express Sur1-Kir6.2 channels (Ortega et al., 2012). In a number of preclinical studies using both transient and permanent models of middle cerebral artery occlusion (MCAO) as well as thromboembolic model of ischemic stroke, it has been shown that treatment with glibenclamide significantly reduces ischemic infarct size and hemispheric swelling, even when administered several hours after the onset of stroke (Simard et al., 2006; Simard et al., 2012; Simard et al., 2009). Analysis of long-term outcomes of glibenclamide treatment have showed marked improvements in sensorimotor and cognitive functions, observed one month after stroke onset (Ortega et al., 2013). The effect of glibenclamide was also examined in rodent models of lethal stroke with malignant cerebral edema, where it significantly reduced edema and mortality rate (Simard et al., 2006; Simard et al., 2010). The molecular basis of the neuroprotective effect of glibenclamide has been attributed to its ability to reduce brain edema through inhibition of Sur1-Trpm4 channels, and to reduce neuroinflammation through inhibition of KATP channel (Kurland et al., 2013). In addition to the preclinical studies, the safety of IV formulation of glibenclamide (BIIB093) has been evaluated in multiple exploratory studies (NCT01132703). Efficacy of IV glibenclamide was also recently evaluated in a randomized, multicenter, prospective, double-blinded, placebo-controlled phase II clinical trial (GAMES-RP) in ischemic stroke patients who were likely to develop malignant edema (NCT01794182). Although the study did not meet the primary efficacy outcome, treatment with IV glibenclamide did show some promising results for measures of edema/swelling and mortality. Midline shift, an imaging marker of neurological deterioration decreased significantly and plasma MMP-9, a biomarker associated with BBB breakdown was lower in patients treated with IV glibenclamide (Kimberly et al., 2018; Sheth et al., 2016; Sheth et al., 2018). Based on this favorable trend from GAMES-RP trial, a phase III randomized, multicenter, double-blinded, placebo controlled, paral-lelgroup, multi-center study (NCT02864953) is currently evaluating the safety and efficacy of this novel anti-edema neuroprotective agent (Pergakis et al., 2019).

MultiStem cells (HLCM051, Athersys) are a distinct subpopulation of adherent human bone marrow cells obtained from heathy adult donors. Prior studies have shown that these adult progenitor cells are not immunogenic, scalable through long-term tissue culture expansion and have the capacity to exert potent immunomodulatory effects in traumatic brain injury, spinal cord injury and stroke (Bedi et al., 2013; Busch et al., 2011; Jacobs et al., 2013; Jellema et al., 2015; Reading et al., 2015; Walker et al., 2012; Yasuhara et al., 2006). Using animal models of acute ischemic stroke, it has also been shown that intravenous administration of Multistem cells 24h after the onset of the insult reduced brain lesion size and neurological deficits, and suppressed the enrichment of ischemia-induced inflammatory genes in the brain and the spleen (Yang et al., 2017a). However, in mice with surgical splenectomy, treatment with Multistem cells following stroke failed to show efficacy, suggesting that a splenic response attenuates pro-inflammatory modulators in the brain to create a regenerative environment for better recovery from stroke (Yang et al., 2017a). However, the potential mchanisms by which cell therapy might improve the outcome after stroke is still not understood. Nevertheless, a phase II, multicenter double-blinded and randomized clinical trial with Multistem cells was carried out for safety assessment and efficacy outcome in stroke patients treated between 24h and 48h after symptom onset and the study outcome has been published. The findings show that a single intravenous administration of Multistem cells (up to 1200 million cells) is safe and well tolerated in stroke patients. But, no significant improvement was observed at 90 days with treatment (Hess et al., 2017). Two additional clinical trials (TREASURE and MASTERS-2) are now enrolling patients, primarily in North America, Europe and Japan to evaluate efficacy of intervention within an earlier time window (<36h).

Imatinib (Gleevec), is a small molecule tyrosine kinase inhibitor that can target both receptor and non-receptor tyrosine kinases and is currently approved by the United States Food and Drug Administration (FDA) for the treatment of cancer. Its most important targets include PDGFRα (platelet derived growth factor receptor α), Flt-3 (FMS-like tyrosine kinase 3), c-fms (CSF-1R; colony-stimulating factor-1-receptor) and Abl tyrosine kinases. The primary goal of the proposed treatment with imatinib in ischemic stroke is to ameliorate the side-effects of thrombolytic treatment with tPA and extend the time window of treatment with tPA. In pre-clinical models of stroke it has been shown that tPA-induced activation of PDGF-CC, a member of the PDGF family and a potent angiogenic factor, upregulates PDGFRα signaling pathway resulting in BBB dysfunction that contributes to infiltration of peripheral immune cells and inflammation (Fredriksson et al., 2004; Su et al., 2008). In these studies, post-ischemic administration of imatinib (1h) along with a delayed tPA treatment (5h) resulted in a significant reduction in hemorrhagic complications and infarct volume (Merali et al., 2015; Su et al., 2008). Inhibition of Flt-3 and c-fms tyrosine kinases by imatinib also alleviate post-stroke inflammatory response through reduction of the pro-inflammatory cytokines IL-1β and TNFα and accumulation of macrophages (Gagalo et al., 2015). A Phase II clinical trial in acute ischemic stroke patients with combined imatinib and intravenous thrombolysis treatment has shown that imatinib is safe and may reduce neurological disability in patients (Wahlgren et al., 2017). Based on these findings, a confirmatory Phase III clinical trial (NCT03639922) is currently underway in Sweden.

Exenatide is a synthetic form of the endogenous glucagon-like-peptide-1 (GLP-1), which is a potent gastrointestinal hormone that regulates blood glucose level by inducing the release of insulin from pancreatic beta cells (Drucker and Nauck, 2006; Lovshin and Drucker, 2009). Studies have shown that GLP-1 receptor is expressed throughout the brain and its expression is upregulated in neurons, astrocytes, microglia and cerebral endothelial cells following an acute ischemic insult (Goke et al., 1995; Hamilton and Holscher, 2009). Increasing evidence indicates that administration of exenatide during or after experimental stroke in mice, rats and gerbils may provide neuroprotection and reduce ischemic infarct volume (Briyal et al., 2012; Lee et al., 2011; Li et al., 2009; Marlet et al., 2018). The efficacy of exenatide in attenuating ischemic brain damage has been observed in both non-diabetic and diabetic animals (Darsalia et al., 2012; Marlet et al., 2018). However, such neuroprotective effect was lost if exenatide is administered 3h after stroke (Teramoto et al., 2011). The proposed mechanism of action of exenatide includes downregulation in the expression of hypoxia-inducible factor-1α (Helton et al., 2005; Jin et al., 2014), reduced expression and activity of inducible nitric oxide synthase (Teramoto et al., 2011), reduced neutrophil infiltration (Kuroki et al., 2016) and decreased ROS generation (Li et al., 2016). Exenatide also reduces expression of MMP-9, an enzyme that plays a significant role in cerebral edema by breaking down the extracellular matrix (Kuroki et al., 2016; Madden, 2012). In spite of the extensive studies in animal models of stroke, clinical trials evaluating the efficacy of exenatide in stroke patients are still sparse. Following a successful pilot study evaluating safety and feasibility of exenatide, a phase II clinical trial (NCT03287076) is currently being conducted to evaluate post-stroke hyperglycemia and improvement in neurological outcome after exenatide treatment for 5 days (Muller et al., 2018).

Poly (ADP-ribose) polymerase-1 (PARP-1), a DNA repair enzyme that is known to be overactivated in brain cells during ischemia causes cellular injury by inducing cellular NAD+ and ATP depletion, mitochondrial dysfunction, reactive oxygen species (ROS) generation, apoptosis inducing factor activation and inflammation (Koh et al., 2004; Moroni, 2008; Strosznajder et al., 2010; Takahashi et al., 1999; Tanaka et al., 2005). Multiple PARP inhibitors have been identified till date that could have diverse effects on ischemia/reperfusion induced brain damage (Berger et al., 2018). Emerging evidence now indicate that JPI-289 (also known as amelparib), a highly-specific, potent and water soluble PARP-1 inhibitor could be a potential neuroprotective agent for treatment of acute ischemic stroke. An ex vivo study in a neuronal culture model of oxygen-glucose deprivation (OGD) has shown that treatment with JPI-289 confers neuroprotection by attenuating OGD-induced depletion of ATP and NAD+ levels, and caspase-3 activation (Kim et al., 2017). Additional studies in rodent models of permanent and transient focal cerebral ischemia have shown that early treatment with JPI-289 can reduce infarct volume and improve functional outcomes by modulating the post-ischemic immune response (Kim et al., 2018; Noh et al., 2018). It has also been reported that the efficacy of various PARP inhibitors in rodent models of stroke is preferentially observed in male mice and is also dependent on the age of the animals (Chen et al., 2020; Eliasson et al., 1997; Hagberg et al., 2004; McCullough et al., 2005; Yuan et al., 2009). However, such gender- and age-dependent differences in the efficacy of JPI-289 for stroke treatment have not been studied yet. A phase I randomized, double-blinded and placebo-controlled clinical trial in Korea has also established the safety and tolerability of multiple doses of JPI-289 in healthy human volunteers (Han et al., 2017). A phase II clinical trial (NCT03062397) is currently being conducted in Korea to evaluate its efficacy in the treatment of ischemic stroke.

Neu2000 is a synthetic derivative of aspirin (acetylsalicylic acid) and the anti-inflammatory drug sulfasalazine (Gwag et al., 2007). It is a dual neuroprotectant targeting both NMDA receptors and free radicals (Cho et al., 2010). Ex vivo studies have shown that Neu2000 is a moderate, uncompetitive and reversible NMDA receptor antagonist with NR2B-subtype specificity and fast binding kinetics (Gwag et al., 2007; Noh et al., 2009). In addition it also blocks free radical injury as a potent cell permeable spin trapping molecule (Cho et al., 2010; Gwag et al., 2007). Randomized and blind experiments in pre-clinical models of transient ischemic stroke have also established the efficacy of Neu2000 in reducing infarct size and improving long-term neurological functions, when administered within 8h of reperfusion (Gwag et al., 2007). In permanent occlusion models, dual injection of Neu2000 at 2h and 16h after the onset of stroke also resulted in significant attenuation of infarct volume (Lee et al., 2007). A phase I double-blinded and randomized clinical trial in a small group of young and elderly volunteers have now established the safety, tolerance and pharmacokinetics of Neu2000 (Cho et al., 2009). A phase II multi-center, randomized and double-blinded clinical trial (NCT02831088) is currently focusing on the efficacy and safety of this neuroprotectant (Hong et al., 2018).

Remote ischemic conditioning (RIC) is based on the concept that repetitive and transient mechanical obstruction of blood vessel at a limb distal from the brain would initiate signals that could increase blood flow to the brain and trigger anti-oxidative, anti-inflammatory, and mitochondria modulatory protective mechanisms in the brain (Hess et al., 2015; Yang et al., 2019b; Zhou et al., 2018). Multiple pre-clinical studies have demonstrated the efficacy of RIC in reducing infarct size and improving functional outcome in rodent models of ischemic stroke (Doeppner et al., 2018; Liu et al., 2014b; Malhotra et al., 2011; Ren et al., 2009; Sun et al., 2012; Wei et al., 2012). In these studies, RIC was induced either by femoral artery occlusion or by manual inflation of a blood pressure cuff on the hindlimb and adminstration of RIC prior to, during, or after stroke were all effective in reducing post-stroke infarct size and brain injury. RIC therapy has also been shown to reduce mortality with intravenous thrombolysis treatment and is effective in both sexes (Hoda et al., 2014; Hoda et al., 2012). However, the exact mechanisms of neuroprotection with RIC therapy are yet unknown. It has been postulated that both circulating mediators and neural mechanisms have central roles in RIC-mediated neuroprotection (Hess et al., 2015; Yang et al., 2019b). In addition to the preclinical evidence, feasibility and safety of RIC as well as the beneficial effects of RIC in ischemic stroke have been observed in several early stage clinical studies (England et al., 2017; Hougaard et al., 2014; Zhao et al., 2018). However, a more recent pilot study in a multicenter randomized trial showed that RIC during or after reperfusion did not reduce brain infarction at 24h after symptom onset (Pico et al., 2020). Several ongoing (Table 2) and recently completed (SERICT-AIS, ReCAST-2) clinical trials are now focusing on the benefits of conditioning following single application of RIC in a prehospital setting or repeated application of RIC for the first 1–2 weeks after an ischemic insult in stroke patients.

6. NINDS/NIH INITIATIVE FOR THE DEVELOPMENT OF NEUROPROTECTIVE STROKE THERAPIES

To accelerate the progress of translational studies for acute neuroprotection in stroke, the National Institute of Neurological Disorders and Stroke (NINDS) in the USA recently announced support for the Stroke Preclinical Assessment Network (SPAN; RFA-NS-18–034). The primary goal of this new approach is to consider highly promising neuroprotective agents or interventions as adjunctive therapies along with or prior to endovascular thrombectomy, to extend the time window for additional intervention and improve functional outcomes. Specifically, six promising therapeutic agents that have been approved by U.S. Food and Drug Administration for treatment in other diseases have been selected for evaluation of efficacy in stroke treatment. These agents will be evaluated simultaneously at multiple sites in experimental animal models of stroke with comorbidities and advanced age in both sexes. These multi-center double-blinded preclinical trials could accelerate the identification of the most promising neuroprotective approaches for future clinical trials. In this section we have summarized the existing literature on these selected neuroprotective agents / interventions (Table 3).

Table 3:

Evaluation of potential neuroprotectants through Stroke Preclinical Assessment Network (SPAN) in USA

TreatmentMechanismsReferences
FasudilA Rho-kinase inhibitor that reduces peripheral immune cell infiltration and oxidative stress. It also enhances cerebral blood flow.Fukuta et al., 2016 Shibuya et al., 2005 Chan et al., 2017 Fukuta et la., 2017
FingolimodA sphingosine-1-phosphate receptor inhibitor that ameliorates peripheral immune cell infiltration and oxidative stress.Li et al., 2017 Dang et al., 2020 Shang et al., 2020 Chamorro et al., 2016 Tian et al., 2018
Remote Ischemic Conditioning (RIC)A series of transient occlusion and reperfusion of blood vessels at a limb distal to the brain triggers neuroprotective effects in the brain. Exact mechanisms are unknown.Ren et al., 2011 Malhotra et al., 2011 Wei et al., 2012 Sun et al., 2012 Doeppner et al., 2018
TocilizumabAn immunosuppressive drug that ameliorates immune response by blocking IL-6 receptor.Wang et al., 2016 Hubodenko et al. 2017 Hubodenko et al., 2019
Uric acidAn antioxidant that suppresses ROS generation, lipid peroxidation and elevates nuclear expression of Nrf2Aamaro et al., 2019 Chamorro et al., 2004 Ya et al., 2018 Valero-Aliena et al., 2018 Dhanesha et al., 2018 Laredo et al., 2016
VeliparibPARP-1 inhibitor that can regulate NAD+ and ATP depletion, mitochondrial dysfunction and immune responseHamby et al., 2007 Takahashi et al., 1997 Berger et al., 2018

Fasudil (AT877 or HA-1077) is an inhibitor of Rho-kinase (ROCK), a downstream target of the small GTP-binding protein Rho. Activation of Rho/ROCK signaling pathway is substantially involved in the pathogenesis of coronary and cerebral vasospasm (Sato et al., 2000; Shimokawa et al., 1999). ROCK activation is also involved in all stages of the inflammatory process (Dong et al., 2010). Thus ROCK inhibitors like fasudil are potential targets for the the treatment of various cardiovascular and central nervous system (CNS) disorders. Fasudil is approved in Japan since 1995 for treatment of cerebral vasospasm. However, it is not yet approved by the FDA for treatment in USA. Fasudil has also shown promise in the attenuation of ischemic brain damage in animal models of MCAO, particularly by decreasing inflammatory response through reduced neutrophil and monocyte infiltration, reduced oxidative damage and improving regional cerebral blood flow (Mueller et al., 2005; Rikitake et al., 2005; Satoh et al., 1996; Satoh et al., 1999; Satoh et al., 2002). However, in spontaneously hypertensive rats treatment with fasudil failed to attenuate ischemic brain damage (Chan and Cipolla, 2017). The safety and usefulness of fasudil in the treatment of stroke patients has also been evaluated in phase III clinical trails (Shibuya et al., 2005). It has been reported that the trial was terminated because of less than anticipated clinical efficacy of the drug (Fukuta et al., 2016). The lack of optimal efficacy of fasudil in clinical trials has been attributed to its short half-life in circulation and poor ability to penetrate the BBB (Mueller et al., 2005). To enhance the bioavailability of fasudil in the brain, in a novel approach, fasudil was encapsulated in the water phase of PEGylated liposomes (liposomal fasudil). Intravenous administration of liposomal fasudil allowed increased release of fasudil in the ischemic region resulting in suppression of neutrophil infiltration and brain cell damage in the MCAO rat models (Fukuta et al., 2016). Combination therapy with tPA in a photochemically induced thrombosis model of stroke, where fasudil was administered prior to tPA treatment also showed decreased intracerebral hemorrhage and increased protection as compared with tPA treatment along with free fasudil or separate treatment with each drug (Fukuta et al., 2017). These findings suggest that liposomal fasudil treatment prior to tPA could reduce the risk of tPA-derived cerebral hemorrhage and extend the time window of tPA treatment. This also implies that liposomal drug delivery system could be a better alternative for stroke therapy with fasudil.

Fingolimod (FTY720) is derived from an atypical amino acid myriocin produced from the fungus Isaria sinclairii, otherwise known as winter-insect and summer plant that is commonly used in traditional Chinese medicine. It is an orally bioavailable lipophilic drug that readily crosses the BBB (Tamagnan et al., 2012) and has been approved by the FDA in 2018 for use in both adolescent and adult patients suffering from multiple sclerosis. In rodent models of transient ischemia, fingolimod has been shown to reduce infarct size and neurological deficits, when administered either before or after the insult (Liu et al., 2013). Fingolimod is structurally similar to the membrane lipid sphingosine (Paugh et al., 2003) and the phosphorylated and active form of fingolimod (fingolimod-phosphate) is analogous to sphingosine-1-phosphate (S1P). The mechanism of action of fingolimod involves its binding to S1P receptors present on lymphocytes causing degradation of the receptor, thereby preventing lymphocyte release from lymphoid tissue (Pham et al., 2008). This decrease in circulating lymphocytes following treatment with fingolimod reduces post-stroke lymphocyte infiltration in the brain resulting in attenuation of post-ischemic inflammatory response and neurotoxicity (Czech et al., 2009; Kraft et al., 2013; Liesz et al., 2011a; Wei et al., 2011). In addition, fingolimod enhances neuroprotection through protein kinase B mediated phosphorylation and inactivation of FOXO3a, a transcription factor that causes oxidative stress (Safarian et al., 2015). The safe use of this drug in patients has already been established in clinical trials involving multiple sclerosis patients, although administration of fingolimod in stroke patients resulted in sustained lymphopenia that persisted for more than 7 days and resolved by day 30 (Fu et al., 2014; Willis and Cohen, 2013). A non-randomized clinical trial in a small group of ischemic stroke patients administered with three oral doses of fingolimod (0.5 mg daily) showed no significant decrease in lesion volume at day 7 compared to placebo treated group (Fu et al., 2014). However, beyond day 7, lesion volume did not increase any further in the fingolimod treated group, although lesion volume continued to increase in the placebo treated group. Significant improvement was also observed in neurological function with fingolimod treatment throughout the 90-day period of the study. Another small randomized study assessed the efficacy of fingolimod when administered prior to treatment with tPA (Zhu et al., 2015). Such combination therapy decreased lesion volume, reduced hemorrhagic transformation and improved functional outcome when compared to tPA treatment alone, suggesting a beneficial role of fingolimod both with and without tPA.

RIC or remote ischemic conditioning has been discussed in more details above (section 5). It involves a series of brief mechanical occlusions and reperfusions of blood vessels at a limb distal to the brain, and exerts its effects through activation of endogenous neuroprotective mechanisms (Ma et al., 2017). This approach is currently being evaluated in multiple ongoing clinical trials in addition to the preclinical studies through the SPAN initiative.

Tocilizumab is a humanized monoclonal antibody against the interleukin-6 (IL-6) receptor. It is an immunosuppressive drug that inhibits the pro-inflammatory effects of IL-6 and has been approved by the FDA for the treatment of autoimmune disease, such as rheumatoid arthritis (Rubbert-Roth et al., 2018; Tanaka et al., 2014). In the classical IL-6 signaling pathway, binding of IL-6 to its receptor activates the Janus-activated kinase / signal transducer and activator of transcription (JAK/STAT) pathway resulting in phosphorylation and subsequent activation STAT3, which then translocates to the nucleus and upregulates genes responsible for inflammation and apoptosis (Scheller et al., 2011; Wang and Sun, 2014). As higher serum levels of IL-6 in stroke patients correlate with larger infarct size and worsening functional outcome (Smith et al., 2004; Waje-Andreassen et al., 2005), there is substantial interest in evaluating the efficacy of treatment with tocilizumab on stroke outcome. However, the effect of IL-6 inhibition on the pathogenesis of ischemic stroke remains controversial. Intravenous administration of anti-IL-6 receptor antibody after the onset of transient MCAO has been shown to exacerbate ischemic brain injury at 24h of reperfusion (Yamashita et al., 2005). Consistent with this finding, intracerebro-ventricular injection of recombinant IL-6 has been shown to reduce early ischemic brain damage in an animal model of permanent MCAO (Loddick et al., 1998). In a separate group of studies, evaluation of the ischemic infarct size between wild-type and IL-6 knockout mice showed that acute stroke outcome (24h) did not differ between the genotypes (Clark et al., 2000; Gertz et al., 2012; Herrmann et al., 2003). The study by Gertz et al (2012) further demonstrated an exacerbation of ischemic lesion volume and worsening of sensori-motor function in IL-6 knockout mice after 4 weeks. In contrast, a study where IL-6 receptor mediated signaling was inhibited with tocilizumab administered 4h after the onset of MCAO in mice showed significant reduction in infarct size, improvement in sensorimotor function and attenuation of microglial activation (Hudobenko et al., 2017). A second study in aged male and female mice have further indicated that amelioration of transient MCAO-induced ischemic brain damage, mortality and behavioral deficit by tocilizumab treatment requires sex-specific dosing, with a 5-fold higher dose requirement for females (Hudobenko et al., 2019).

Uric acid, a product of purine metabolism is a powerful endogenous antioxidant in the circulation (Pasalic et al., 2012). However, high serum uric acid levels (hyperuricemia) is neurotoxic as a pro-oxidant and may increase the risk of stroke morbidity and mortality (Chamorro and Planas, 2004; Kanellis and Johnson, 2003; Li et al., 2014; Li et al., 2015). Despite this controversy, earlier studies have shown that physiologically relevant concentrations of uric acid confers neuroprotection to cultured neurons from glutamate-induced (excitotoxic) or peroxynitrite-induced (nitrosative) cell death by scavenging ROS and suppressing lipid peroxidation (Gursoy-Ozdemir et al., 2004; Justicia et al., 2017; Romanos et al., 2007; Squadrito et al., 2000). The neuroprotective effects of uric acid has also been attributed to its role in elevating nuclear expression of nuclear factor E2-related factor 2 (Nrf2) protein, an essential transcription factor that regulates the transcription of multiple antioxidant and detoxifying genes (Kanninen et al., 2015; Nakka et al., 2016; Ya et al., 2018). In animal models of transient ischemic insult, exogenous administration of uric acid has been shown to attenuate ischemic brain damage and behavioral deficits by reducing lipid, protein and DNA peroxidation as well as post-ischemic infiltration of neutrophils (Chamorro et al., 2014; Justicia et al., 2017; Onetti et al., 2015; Romanos et al., 2007; Ya et al., 2018; Yu et al., 1998). The efficacy of uric acid in reducing stroke-induced brain damage was more pronounced following cotreatment with tPA, suggesting that combination therapy extends the benefits of tPA treatment alone (Romanos et al., 2007). In animal studies, uric acid treatment was also effective in improving stroke outcome in both males and females (Aliena-Valero et al., 2018; Dhanesha et al., 2018). In a randomized, double-blinded, placebo-controlled, phase III clinical trial (URICO-ICTUS), uric acid has been shown to be safe and improve outcomes in stroke patients when administered along with tPA, as well as in patients receiving both intravenous tPA and mechanical thrombectomy (Amaro et al., 2016; Amaro et al., 2019; Chamorro et al., 2017; Chamorro et al., 2004).

Veliparib (ABT-888) is another PARP-1 inhibitor that has similar mechanism of action as JPI-289 (amelparib) described above (section 5). Veliparib has good oral bioavailability, can cross the BBB and is well tolerated in cancer patients (Donawho et al., 2007; Reck et al., 2017). It has been granted “orphan drug designation” by FDA in 2016 for treatment of advanced squamous non–small cell lung cancer. However, its efficacy in attenuating ischemic brain damage in preclinical animal models of stroke or clinical trials in stroke patients has not been evaluated yet.

7. EMERGING NEUROPROTECTANTS FOR STROKE TREATMENT

It is now expected that development of future strategies for stroke therapy would require novel approaches for selection of neuroprotective agents. Such approaches should emphasize on agents that can readily cross the BBB as well as target the upstream components or multiple aspects of the ischemic cascade. In this section we have evaluated the therapeutic potential of two emerging peptide-based agents, a pharmacological compound and several epigenetic factors as future neuroprotective strategies to reduce the impact of ischemic brain injury (Table 4).

Table 4:

Emerging approaches for neuroprotection in ischemic stroke

TreatmentMechanismsReferences
AdropinAmeliorates neurovascular dysfunction to confer neuroprotectionYang et al., 2020 Shahjouei et al., 2016 Altintas et al, 2016

STEP-mimeticTargets excitotoxic signaling cascade downstream of NMDARs to confer neuroprotectionPoddar et al., 2019 Deb et al., 2013 Poddar et al 2010

VerapamilReduces excitotoxic damage as a L- type calcium channel blocker (and vasodilator)Fraser et al., 2017; Maniskas et al., 2016

Micro RNAs:
microRNA-29aReduces excitotoxicity by preserving astrocytic glutamate transporter-1 levelOuyang et al., 2013 Tuo et al., 2017



microRNA-223Reduces excitotoxicity by regulating expression of NR2B-NMDAR and GluR2-AMPARHarraz et al., 2012



microRNA-424Reduces oxidative stress by enhancing expression of SOD, MnSOD and Nrf2Zhao et al., 2013 Liu et al., 2015



microRNA-99aReduces oxidative stress and neuronal apoptosisTao et al., 2015; Zhao et al., 2017



microRNA-23a-3pReduces oxidative stress by enhancing expression of MnSOD and reducing nO and 3-NT productionZhao et al., 2014



microRNA-106b-5p (antagomir)Reduces oxidative stress by restoring SOD activity and increasing the expression of Mcl-1Li et al., 2015, 2017



microRNA-124Reduces inflammation by promoting anti-inflammatory M2 phenotype of microglial/macrophageTaj et al., 2016 Sun et al., 2013



microRNA-let-7c-5pReduces inflammation by inhibiting microglial activationNi et al., 2015



microRNA-181a (antagomir)Reduces inflammation by inhibiting NF- kB activation, microglial activation, leukocyte infiltration and decrease in level of GRP78, an ER stress regulatorOuyang et al., 2012 Xu et al., 2015



microRNA 15a/16-1 (antagomir)Reduces BBB dysfunction, inflammatory response and apoptosisYang et al., 2017 Ma et al., 2020

Adropin is a recently identified small polypeptide (4.9 kDa) encoded by the Energy homeostasis gene (Enho), which is expressed in multiple organs including the brain (Aydin et al., 2013; Kumar et al., 2008; Wong et al., 2014). Multiple functions of adropin includes its ability to regulate glucose and lipid homeostasis and insulin sensitivity (Aydin, 2014; Kumar et al., 2008). It also enhances mitochondrial function and plays a role in fatty acid oxidation (Gao et al., 2015). Furthermore, adropin has beneficial effects on endothelial cell function by reducing permeability through activation of endothelial nitric oxide synthase (eNOS) signaling pathways (Lovren et al., 2010). Emerging evidences indicates that adropin level is significantly downregulated in rat brain microvascular endothelial cells exposed to hypoxia and low glucose, resulting in increased paracellular permeability, while treatment with synthetic adropin attenuates the increased endothelial permeability (Yang et al., 2016). This would imply that adropin could be protective against endothelial barrier dysfunction during ischemia. Consistent with the above finding, more recent studies in animal models of cerebral ischemia have shown significant reduction in adropin level, which is accompanied with a larger infarct size. Treatment with synthetic adropin reduced infarct size through eNOS activation and reduced oxidative damage (Yang et al., 2017b; Yang et al., 2020). The role of adropin-mediated activation of eNOS signaling pathway in neuroprotection against cerebral ischemia was further confirmed using adropin overexpressing transgenic mice (AdrTg), adropin knockout mice (Enho−/−) and eNOS knockout mice (eNOS−/−) subjected to cerebral stroke. This later study showed significant reduction in ischemic brain damage in AdrTg mice when compared to wild-type control. Conversely, adropin deficient mice had larger strokes compared to wild-type littermate controls. Pharmacological treatment with synthetic adropin significantly reduced infarct size and protected against stroke-induced BBB damage. This protective effect of adropin was completely abolished in eNOS−/− mice (Yang et al., 2020), suggesting that eNOS is involved in the underlying mechanisms of protection by adropin in stroke. Understanding the long-term effects of adropin treatment on stroke outcomes and the underlying signaling mechanism(s) could therefore offer a novel therapeutic strategy for the treatment of ischemic brain damage.

The tyrosine phosphatase STEP, also known as Ptpn5, is an intracellular phosphatase that is expressed specifically in the central nervous system (Boulanger et al., 1995; Lombroso et al., 1993), and whose specific targets and functions are beginning to emerge. STEP is expressed in the neurons of the striatum, cortex, hippocampus and related structures, and its substrate affinity is regulated through phosphorylation/dephosphorylation of a serine residue in the conserved regulatory domain termed the KIM (kinase interacting motif) domain (Bult et al., 1997; Paul et al., 2003; Paul et al., 2000; Poddar et al., 2010; Pulido et al., 1998). STEP can dephosphorylate and inactivate several key neuronal signaling proteins that includes the extracellular signal-regulating kinase 1 and 2 (ERK1/2), stress activated protein kinases p38, the Src family tyrosine kinases Fyn and Pyk2, NR2B-subunit of NMDARs and GluA2 and GluA3 subunit of AMPA receptors (Nguyen et al., 2002; Paul et al., 2003; Poddar et al., 2010; Poddar et al., 2016; Snyder et al., 2005; Won et al., 2019; Xu et al., 2012). Aberrant increase in the activation of these proteins during an ischemic insult plays a role in excitotoxicity, oxidative stress and inflammatory response. As an inhibitor of the activity of these proteins, STEP has the potential to target multiple aspects of the ischemic cascade to confer neuroprotection. Indeed, in cell culture models of excitotoxicity and oxygen glucose deprivation, dephosphorylation and subsequent activation of STEP has been shown to contribute to neuroprotection (Deb et al., 2013; Poddar et al., 2010). Using a rat model of transient focal ischemia, it has also been shown that rapid activation of STEP during the ischemic insult provides initial neuroprotection, while degradation of active STEP over time leads to secondary activation of deleterious processes, resulting in progression of ischemic brain damage (Poddar et al., 2010). Another potentially important finding is that STEP activity decreases with age-associated increase in oxidative stress (Deb et al., 2011; Rajagopal et al., 2016), suggesting that the loss of this protective response may be a contributing factor for the increased susceptibility of the aging brain to ischemic brain damage (Howard et al., 1987; Nakayama et al., 1994). Consistent with this interpretation, studies in STEP knockout (KO) mice further showed that the loss of endogenous STEP leads to exacerbation of ischemic brain injury as observed 24h after a mild ischemic insult. Based on these observations, additional studies developed a novel brain-permeable STEP-derived peptide (STEP-mimetic) that is resistant to degradation (Deb et al., 2013). Intravenous administration of this peptide in a rat model of ischemic stroke was not only effective in limiting stroke injury and functional deficits but also facilitated long-term recovery, even when administered 6h after the onset of the insult (Deb et al., 2013; Poddar et al., 2019). Studies in STEP KO mice further showed that restoration of the STEP signaling pathway with the administration of the STEP-mimetic could also attenuate the exacerbation of ischemic brain injury in the absence of endogenous STEP (Deb et al., 2013). These findings highlight the importance of STEP in neuroprotection against ischemic brain injury. Establishing the efficacy of the STEP-mimetic in attenuating ischemic brain damage in animals models with comorbidities and understanding the molecular basis of this neuroprotection would help promote the STEP-mimetic as a novel therapeutic strategy for stroke treatment.

Verapamil is a phenylalkylamine and a member of the non-dihydropyridine class of calcium channel blockers (Bergson et al., 2011). It is primarily a L-type calcium channel blocker with higher affinity for depolarized channels rather than resting channels, which prevents the contraction of vascular smooth muscle cells causing vasodilation throughout peripheral circulation thus alleviating angina (Bergson et al., 2011). Verapamil is currently approved by the FDA for the treatment of hypertension, angina and arrhythmia (Frishman and Charlap, 1983; McTavish and Sorkin, 1989). Intra-arterial verapamil administration is also thought to be effective for treatment cerebral vasopasm, a common complication of aneurysmal subarachnoid hemorrhage (Keuskamp et al., 2008; Mikeladze et al., 2013; Stuart et al., 2011). Earlier studies on the application of verapamil for stroke treatment in animal models have mixed outcome depending on the animal model of stroke and the post-stroke time of intervention (Berger et al., 1984; Roy et al., 1985; Ueda et al., 1989; Wauquier et al., 1989). However, in more recent studies intra-arterial administration of verapamil following recanalization in a mouse model of focal cortical ischemia showed significant reduction in infarct volume and improvement in functional outcome (Fraser et al., 2017; Maniskas et al., 2016). Most importantly, these studies also showed that verapamil treatment did not have any effect on cerebral perfusion, implying that the protective mechanisms of action in stroke is independent from its vasodilatory effects. Although the exact mechanism of action of verapamil in stroke treatment is still not understood the authors postulated that through its inhibition of L-type calcium channels verapamil could reduce excitotoxic damage. Further support for this hypothesis comes from in vitro studies where verapamil administration to neurons in culture following OGD prevented neuronal injury in a dose-dependent manner (Fraser et al., 2017). In the same study, a phase I trial (SAVER-I) evaluating the feasibility and safety of intra-arterial administration of verapamil in human patients showed that at a clinically applicable dose range, intra-arterial administration of verapamil following thrombectomy is safe and feasible. These findings not only establish the feasibility of developing of verapamil as a new neuroprotectant for stroke therapy but also highlight the importance of intra-arterial delivery of neuroprotectants for stroke treatment.

The neuroprotectants identified till date for stroke therapy are involved in regulating intracellular signaling pathways to reduce post-stroke edema, excitotoxicity, oxidative stress, inflammation and apoptosis. In addition to pursuing these conventional therapeutic approaches current attention has shifted towards the identification of epigenetic factors that might influence the etiology and progression of stroke (Ng et al., 2018). MicroRNAs (miRNAs) are emerging as an important player in the field of epigenetics and are involved mainly with message translation rather than with gene transcription. They are the best characterized sub-class of non-coding RNAs and the sequences coding for miRNAs often arise from intronic DNA and regulate the gene products coded by adjacent exons. They can regulate mRNA translation by binding to and retarding or accelerating their degradation. In addition, miRNA binding to mRNA can block message translation (Krupinski and Slevin, 2013). MicroRNAs appears to modulate different aspects of stroke pathophysiology, including excitotoxicity, oxidative stress, inflammation and BBB dysfunction, and several miRNAs that are neuroprotective tend to be downregulated in stroke, while miRNAs that could promote neurotoxic signaling tend to be upregulated (Khoshnam et al., 2017; Ng et al., 2018). With the development of synthetic miRNAs that could either mimic the function of biological miRNAs (agomirs) or inhibit the function of specific miRNAs (antagomirs), it is now possible to evaluate the efficacy of upregulating or downregulating miRNA levels in promoting neuroprotection in stroke. Based on the current literature, here we have highlighted the role of several promising miRNAs as potential neuroprotective agents in cerebral ischemic stroke (Table 4).

Post-ischemic down regulation of the glutamate transporter-1 (GLT-1) has been associated with the accumulation of glutamate and neuronal excitotoxicity. It has now been shown that expression of miR-29a and miR-223 are involved in regulating post-ischemic excitotoxicity and cell death. Intravenous administration of miR-29a agomir following reperfusion protects astrocytes and preserves astrocyte GLT-1 level by targeting the pro-apoptotic protein PUMA (p53 upregulated modulator of apoptosis). Overexpression of mir-223 also controls neuronal injury by reducing the levels of the GluR2 subunit of AMPA receptors and NR2B-subunit of NMDA receptors (Harraz et al., 2012; Ouyang et al., 2013; Tuo et al., 2017).

Studies have also shown that miR-424, miR-99a, miR-23a-3p agomirs as well as miR-106b-5p antagomir protects against ischemic brain injury by reducing oxidative stress. miR-424 levels has been shown to decrease in plasma of patients with acute ischemic stroke as well as in plasma and ipsilateral brain tissue of mouse after ischemic stroke. Accordingly, treatment with miR-424 decreases cerebral infarct volume by increasing the expression and activation of superoxide dismutase (SOD), manganese SOD (MnSOD) and the redox sensitive transcription factor Nrf2. It also suppresses microglial activation and TNFα production (Liu et al., 2015; Zhao et al., 2013). The level of miR-99a, a suppressor of tumorigenicity, has also been observed to be significantly downregulated in the plasma of acute ischemic stroke patients. Furthermore, the neuroprotective role of miR-99a was demonstrated in a mouse model of transient MCAO, where intracerebroventricular injection of miR-99a agomir immediately after MCAO reduced both infarct volume and neural apoptosis (Li et al., 2011; Sun et al., 2011a; Tao et al., 2015). Mechanistic studies further showed that miR-99a can alleviate oxidative stress injury and promote neural survival following stroke. Similarly, another suppressor of tumorigenicity miR23a-3p has been shown to reduce ischemic infarction volume and lessen oxidative stress when miR23a-3p agomir is injected intracerebroventricularly immediately after MCAO (Chen et al., 2019; Ding et al., 2019; Zhao et al., 2014). On the other hand, miR-106b-5p, which functions as a tumor promoter has also been reported to be a major contributor of oxidative stress and cerebral ischemic injury (Li et al., 2017a; Liu et al., 2014a; Xiang et al., 2015). Furthermore, miR-106b-5p antagomir has been shown to significantly decrease ischemic infarct volume and neurological deficits. At the mechanistic level, miR-106b-5p antagomir reduced malondialdehyde content, restored SOD activity, increased the expression of Mcl-1, an anti-apoptotic member of the Bcl-2 family, and decreased the expression of Bax in the ischemic cortex (Li et al., 2017a).

Micro RNAs that have been implicated in the regulation of inflammation during stroke includes miR-124, miR-let-7c-5p and miR-181a. miR124 is almost exclusively expressed in the CNS (Lagos-Quintana et al., 2002; Landgraf et al., 2007). Post-ischemic administration of miR-124 has been shown to reduce infarct size at 24h after MCAO by modulating polarization of activated microglia and infiltrating macrophages towards the anti-inflammatory M2 phenotype (Hamzei Taj et al., 2016; Sun et al., 2013). miR-let-7c is another important regulator of macrophage polarization (Banerjee et al., 2013). Recent studies have shown that the level of miR-let7c-5p, a member of the let-7c family, is significantly decreased in the plasma of patients with ischemic stroke as well as in the plasma and ipsilateral brain tissue of mice subjected to MCAO/reperfusion (Ni et al., 2015). The study further showed that intracerebroventricular injection of miR-let7c-5p agomir decreased the infarct volume through inhibition of microglial activation and attenuated neurological deficits (Ni et al., 2015). Expression of miR-181a, which is enriched in the brain, has been shown to increase in the ischemic core following stroke and was associated with decrease in the level of GRP78, a master regulator of endoplasmic reticulum stress with antiapoptotic properties. Conversely, reducing the level of miR-181a with an antagomir reduced ischemic injury and was associated with increase in GRP78 level (Lee, 2005; Miska et al., 2004; Ouyang et al., 2012). Additional studies further showed that the reduction in brain infarct size following post-stroke treatment with miR-181a antagomir was associated with reduced NF-κB activation, microglial activation and leukocyte infiltration (Xu et al., 2015). The findings imply that reducing or blocking miR-181a protects the brain from both oxidative stress and inflammatory response following ischemia.

The role of microRNAs in BBB dysfunction following ischemic stroke is also evident from emerging studies. Two highly conserved microRNAs, miR-15a and miR-16–1 binds to common mRNA targets forming a structural and functional cluster termed as the miR15a/16–1 cluster (Calin et al., 2008; Klein et al., 2010). The cluster was first identified in human leukemia (Calin et al., 2008) and more recently dysregulation of miR-15a/16–1 levels in plasma of stroke patients has been reported (Tan et al., 2009). Subsequent studies in animal models of stroke showed significant increase in the level of both miR-15a and miR-16–1 in the ipsilateral hemisphere (Yang et al., 2017d). Utilizing miR-15a/16–1 knockout mice and pharmacological intervention with miR15a/16–1 antagomirs, the study further established that miR15a/16–1 in the brain negatively regulates the outcome of stroke by promoting inflammatory response and inhibiting the antiapoptotic genes bcl-2 and bcl-w. In an additional study, experimentally induced stroke in mice with an endothelial cell selective deletion of miR15a/16–1 showed a smaller infarct size, reduced BBB leakage and decreased infiltration of peripheral immune cells (Ma et al., 2020). Together, the above findings not only expand our understanding of the roles of miRNAs in stroke, but also identify a new direction for the development of future neuroprotective strategies.

8. CONCLUSION

It is evident from the existing literature that a large number of therapeutic agents have either undergone clinical trials or currently being assessed in pre-clinical studies for evaluation of efficacy in acute ischemic stroke. In spite of this extensive effort, the development of clinically effective neuroprotective agents is still elusive. Stroke is a complex disorder and our current understanding of the mechanistic basis indicates simultaneous and sequential activation of multiple detrimental signaling cascades in both the core and the penumbra. Moreover, with the advent of new technologies such as proteomics, metabolomics and RNA sequencing, new molecular events involved in ischemic brain damage are still emerging. Development of therapeutic agents that target a single event in the ischemic cascade may not be effective in spite of successful inhibition of the specific target. It is therefore essential to focus primarily on pleiotropic, multi-target agents that could intervene at multiple levels of the ischemic cascade to confer neuroprotection. However, identification of such agents could be a daunting task and could also be impossible. In addition, toxicity of such pleiotropic drugs could also be an issue. A clinically useful alternative could therefore be the development of combinational therapy. This would include multiple therapeutic compounds that could target both complementary and synergistic pathways in the acute stage, based on our knowledge of the distinct temporal profile of the progression of ischemic cascade. Development of such optimal therapies by combining neuroprotective agents that are currently undergoing clinical/pre-clinical assessment holds promise and it is only a matter of time before further advancement in stroke research yields a breakthrough.

ACKNOWLEDGMENT

Authors are supported by grants from the National Institute of Neurological Disorders and Stroke, NINDS/NIH (R01 NS059962 to S.P and R01 NS103094 and R01 NS109816 to E.C.J.).

Footnotes

DECLARATION OF COMPETING INTERESTS

The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES:

  • Aarts M, Liu Y, Liu L, Besshoh S, Arundine M, Gurd JW, Wang YT, Salter MW, Tymianski M, 2002. Treatment of ischemic brain damage by perturbing NMDA receptor- PSD-95 protein interactions. Science 298, 846–850. [Abstract] [Google Scholar]
  • Abdoulaye IA, Guo YJ, 2016. A Review of Recent Advances in Neuroprotective Potential of 3-NButylphthalide and Its Derivatives. Biomed Res Int 2016, 5012341. [Europe PMC free article] [Abstract] [Google Scholar]
  • Al-Kuraishy HM, Al-Gareeb AI, Naji MT, Al-Mamorry F, 2020. Role of vinpocetine in ischemic stroke and poststroke outcomes: A critical review. Brain Circ 6, 1–10. [Europe PMC free article] [Abstract] [Google Scholar]
  • Albers GW, Goldstein LB, Hess DC, Wechsler LR, Furie KL, Gorelick PB, Hurn P, Liebeskind DS, Nogueira RG, Saver JL, Consortium SV, 2011. Stroke Treatment Academic Industry Roundtable (STAIR) recommendations for maximizing the use of intravenous thrombolytics and expanding treatment options with intra-arterial and neuroprotective therapies. Stroke 42, 2645–2650. [Abstract] [Google Scholar]
  • Aliena-Valero A, Lopez-Morales MA, Burguete MC, Castello-Ruiz M, Jover-Mengual T, Hervas D, Torregrosa G, Leira EC, Chamorro A, Salom JB, 2018. Emergent Uric Acid Treatment is Synergistic with Mechanical Recanalization in Improving Stroke Outcomes in Male and Female Rats. Neuroscience 388, 263–273. [Abstract] [Google Scholar]
  • Amarenco P, Bogousslavsky J, Caplan LR, Donnan GA, Hennerici MG, 2009. Classification of stroke subtypes. Cerebrovasc Dis 27, 493–501. [Abstract] [Google Scholar]
  • Amaro S, Laredo C, Renu A, Llull L, Rudilosso S, Obach V, Urra X, Planas AM, Chamorro A, Investigators U-I, 2016. Uric Acid Therapy Prevents Early Ischemic Stroke Progression: A Tertiary Analysis of the URICO-ICTUS Trial (Efficacy Study of Combined Treatment With Uric Acid and r-tPA in Acute Ischemic Stroke). Stroke 47, 2874–2876. [Abstract] [Google Scholar]
  • Amaro S, Renu A, Laredo C, Castellanos M, Arenillas JF, Llull L, Rudilloso S, Urra X, Obach V, Chamorro A, on behalf of the, U.-I.i., 2019. Relevance of Collaterals for the Success of Neuroprotective Therapies in Acute Ischemic Stroke: Insights from the Randomized URICOICTUS Trial. Cerebrovasc Dis 47, 171–177. [Abstract] [Google Scholar]
  • Amin-Hanjani S, Stagliano NE, Yamada M, Huang PL, Liao JK, Moskowitz MA, 2001. Mevastatin, an HMG-CoA reductase inhibitor, reduces stroke damage and upregulates endothelial nitric oxide synthase in mice. Stroke 32, 980–986. [Abstract] [Google Scholar]
  • Astrup J, Siesjo BK, Symon L, 1981. Thresholds in cerebral ischemia - the ischemic penumbra. Stroke 12, 723–725. [Abstract] [Google Scholar]
  • Aydin S, 2014. Three new players in energy regulation: preptin, adropin and irisin. Peptides 56, 94–110. [Abstract] [Google Scholar]
  • Aydin S, Kuloglu T, Aydin S, Eren MN, Yilmaz M, Kalayci M, Sahin I, Kocaman N, Citil C, Kendir Y, 2013. Expression of adropin in rat brain, cerebellum, kidneys, heart, liver, and pancreas in streptozotocin-induced diabetes. Mol Cell Biochem 380, 73–81. [Abstract] [Google Scholar]
  • Bai J, Lyden PD, 2015. Revisiting cerebral postischemic reperfusion injury: new insights in understanding reperfusion failure, hemorrhage, and edema. Int J Stroke 10, 143–152. [Abstract] [Google Scholar]
  • Balduini W, Carloni S, Cimino M, 2016. Preclinical randomized controlled multicenter trials (pRCT) in stroke research: a new and valid approach to improve translation? Ann Transl Med 4, 549. [Europe PMC free article] [Abstract] [Google Scholar]
  • Balduini W, De Angelis V, Mazzoni E, Cimino M, 2001. Simvastatin protects against long-lasting behavioral and morphological consequences of neonatal hypoxic/ischemic brain injury. Stroke 32, 2185–2191. [Abstract] [Google Scholar]
  • Ballarin B, Tymianski M, 2018. Discovery and development of NA-1 for the treatment of acute ischemic stroke. Acta Pharmacol Sin 39, 661–668. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bamford J, Sandercock P, Dennis M, Burn J, Warlow C, 1991. Classification and natural history of clinically identifiable subtypes of cerebral infarction. Lancet 337, 1521–1526. [Abstract] [Google Scholar]
  • Banerjee S, Xie N, Cui H, Tan Z, Yang S, Icyuz M, Abraham E, Liu G, 2013. MicroRNA let-7c regulates macrophage polarization. J Immunol 190, 6542–6549. [Europe PMC free article] [Abstract] [Google Scholar]
  • Banjara M, Ghosh C, 2017. Sterile Neuroinflammation and Strategies for Therapeutic Intervention. Int J Inflam 2017, 8385961. [Europe PMC free article] [Abstract] [Google Scholar]
  • Banno M, Mizuno T, Kato H, Zhang G, Kawanokuchi J, Wang J, Kuno R, Jin S, Takeuchi H, Suzumura A, 2005. The radical scavenger edaravone prevents oxidative neurotoxicity induced by peroxynitrite and activated microglia. Neuropharmacology 48, 283–290. [Abstract] [Google Scholar]
  • Becker K, Kindrick D, Relton J, Harlan J, Winn R, 2001. Antibody to the alpha4 integrin decreases infarct size in transient focal cerebral ischemia in rats. Stroke 32, 206–211. [Abstract] [Google Scholar]
  • Bedi SS, Hetz R, Thomas C, Smith P, Olsen AB, Williams S, Xue H, Aroom K, Uray K, Hamilton J, Mays RW, Cox CS Jr., 2013. Intravenous multipotent adult progenitor cell therapy attenuates activated microglial/macrophage response and improves spatial learning after traumatic brain injury. Stem Cells Transl Med 2, 953–960. [Europe PMC free article] [Abstract] [Google Scholar]
  • Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, Delling FN, Djousse L, Elkind MSV, Ferguson JF, Fornage M, Jordan LC, Khan SS, Kissela BM, Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino ME, O’Flaherty M, Pandey A, Perak AM, Rosamond WD, Roth GA, Sampson UKA, Satou GM, Schroeder EB, Shah SH, Spartano NL, Stokes A, Tirschwell DL, Tsao CW, Turakhia MP, VanWagner LB, Wilkins JT, Wong SS, Virani SS, American Heart Association Council on, E., Prevention Statistics, C., Stroke Statistics, S., 2019. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation 139, e56–e528. [Abstract] [Google Scholar]
  • Bereczki D, Fekete I, 2008. Vinpocetine for acute ischaemic stroke. Cochrane Database Syst Rev, CD000480. [Europe PMC free article] [Abstract] [Google Scholar]
  • Berger JR, Busto R, Ginsberg MD, 1984. Verapamil: failure of metabolic amelioration following global forebrain ischemia in the rat. Stroke 15, 1029–1032. [Abstract] [Google Scholar]
  • Berger NA, Besson VC, Boulares AH, Burkle A, Chiarugi A, Clark RS, Curtin NJ, Cuzzocrea S, Dawson TM, Dawson VL, Hasko G, Liaudet L, Moroni F, Pacher P, Radermacher P, Salzman AL, Snyder SH, Soriano FG, Strosznajder RP, Sumegi B, Swanson RA, Szabo C, 2018. Opportunities for the repurposing of PARP inhibitors for the therapy of non-oncological diseases. Br J Pharmacol 175, 192–222. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bergson P, Lipkind G, Lee SP, Duban ME, Hanck DA, 2011. Verapamil block of T-type calcium channels. Mol Pharmacol 79, 411–419. [Europe PMC free article] [Abstract] [Google Scholar]
  • Boehme AK, Esenwa C, Elkind MS, 2017. Stroke Risk Factors, Genetics, and Prevention. Circ Res 120, 472–495. [Europe PMC free article] [Abstract] [Google Scholar]
  • Boulanger LM, Lombroso PJ, Raghunathan A, During MJ, Wahle P, Naegele JR, 1995. Cellular and molecular characterization of a brain-enriched protein tyrosine phosphatase. J Neurosci 15, 1532–1544. [Europe PMC free article] [Abstract] [Google Scholar]
  • Braeuninger S, Kleinschnitz C, 2009. Rodent models of focal cerebral ischemia: procedural pitfalls and translational problems. Exp Transl Stroke Med 1, 8. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bratane BT, Cui H, Cook DJ, Bouley J, Tymianski M, Fisher M, 2011. Neuroprotection by freezing ischemic penumbra evolution without cerebral blood flow augmentation with a postsynaptic density-95 protein inhibitor. Stroke 42, 3265–3270. [Abstract] [Google Scholar]
  • Briyal S, Gulati K, Gulati A, 2012. Repeated administration of exendin-4 reduces focal cerebral ischemia-induced infarction in rats. Brain Res 1427, 23–34. [Abstract] [Google Scholar]
  • Bult A, Zhao F, Dirkx R Jr., Raghunathan A, Solimena M, Lombroso PJ, 1997. STEP: a family of brain-enriched PTPs. Alternative splicing produces transmembrane, cytosolic and truncated isoforms. Eur J Cell Biol 72, 337–344. [Abstract] [Google Scholar]
  • Busch SA, Hamilton JA, Horn KP, Cuascut FX, Cutrone R, Lehman N, Deans RJ, Ting AE, Mays RW, Silver J, 2011. Multipotent adult progenitor cells prevent macrophage-mediated axonal dieback and promote regrowth after spinal cord injury. J Neurosci 31, 944–953. [Europe PMC free article] [Abstract] [Google Scholar]
  • Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, Taccioli C, Zanesi N, Garzon R, Aqeilan RI, Alder H, Volinia S, Rassenti L, Liu X, Liu CG, Kipps TJ, Negrini M, Croce CM, 2008. MiR-15a and miR-16–1 cluster functions in human leukemia. Proc Natl Acad Sci U S A 105, 5166–5171. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cekanaviciute E, Fathali N, Doyle KP, Williams AM, Han J, Buckwalter MS, 2014. Astrocytic transforming growth factor-beta signaling reduces subacute neuroinflammation after stroke in mice. Glia 62, 1227–1240. [Europe PMC free article] [Abstract] [Google Scholar]
  • Chamorro A, Amaro S, Castellanos M, Gomis M, Urra X, Blasco J, Arenillas JF, Roman LS, Munoz R, Macho J, Canovas D, Marti-Fabregas J, Leira EC, Planas AM, Investigators U-I, 2017. Uric acid therapy improves the outcomes of stroke patients treated with intravenous tissue plasminogen activator and mechanical thrombectomy. Int J Stroke 12, 377–382. [Abstract] [Google Scholar]
  • Chamorro A, Amaro S, Castellanos M, Segura T, Arenillas J, Marti-Fabregas J, Gallego J, Krupinski J, Gomis M, Canovas D, Carne X, Deulofeu R, Roman LS, Oleaga L, Torres F, Planas AM, Investigators U-I, 2014. Safety and efficacy of uric acid in patients with acute stroke (URICO-ICTUS): a randomised, double-blind phase 2b/3 trial. Lancet Neurol 13, 453–460. [Abstract] [Google Scholar]
  • Chamorro A, Dirnagl U, Urra X, Planas AM, 2016. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol 15, 869–881. [Abstract] [Google Scholar]
  • Chamorro A, Meisel A, Planas AM, Urra X, van de Beek D, Veltkamp R, 2012. The immunology of acute stroke. Nat Rev Neurol 8, 401–410. [Abstract] [Google Scholar]
  • Chamorro A, Planas AM, 2004. Yin and yang of uric acid in patients with stroke. Stroke 35, e1112; author reply e11–12. [Abstract] [Google Scholar]
  • Chamorro A, Planas AM, Muner DS, Deulofeu R, 2004. Uric acid administration for neuroprotection in patients with acute brain ischemia. Med Hypotheses 62, 173–176. [Abstract] [Google Scholar]
  • Chan SL, Cipolla MJ, 2017. Treatment with low dose fasudil for acute ischemic stroke in chronic hypertension. J Cereb Blood Flow Metab 37, 3262–3270. [Europe PMC free article] [Abstract] [Google Scholar]
  • Chao J, Chao L, 2005. Kallikrein-kinin in stroke, cardiovascular and renal disease. Exp Physiol 90, 291–298. [Abstract] [Google Scholar]
  • Chao J, Chao L, 2006. Experimental therapy with tissue kallikrein against cerebral ischemia. Front Biosci 11, 1323–1327. [Abstract] [Google Scholar]
  • Chau R, 2005. Methods and use of motoneuronotrophic factors, US Patent # US6841531B2.
  • Chau R, Ko P, 2007. MNTF peptides and compositions and methods of use, US Patent # US7183373B2.
  • Chen F, Qi S, Zhang X, Wu J, Yang X, Wang R, 2019. miR-23a-3p suppresses cell proliferation in oral squamous cell carcinomas by targeting FGF2 and correlates with a better prognosis: miR-23a-3p inhibits OSCC growth by targeting FGF2. Pathol Res Pract 215, 660–667. [Abstract] [Google Scholar]
  • Chen J, Li X, Xu S, Zhang M, Wu Z, Zhang X, Xu Y, Chen Y, 2020. Delayed PARP-1 Inhibition Alleviates Post-stroke Inflammation in Male Versus Female Mice: Differences and Similarities. Front Cell Neurosci 14, 77. [Europe PMC free article] [Abstract] [Google Scholar]
  • Chen N, Zhou Z, Li J, Li B, Feng J, He D, Luo Y, Zheng X, Luo J, Zhang J, 2018. 3-nbutylphthalide exerts neuroprotective effects by enhancing anti-oxidation and attenuating mitochondrial dysfunction in an in vitro model of ischemic stroke. Drug Des Devel Ther 12, 42614271. [Europe PMC free article] [Abstract] [Google Scholar]
  • Chen RL, Balami JS, Esiri MM, Chen LK, Buchan AM, 2010a. Ischemic stroke in the elderly: an overview of evidence. Nat Rev Neurol 6, 256–265. [Abstract] [Google Scholar]
  • Chen ZB, Huang DQ, Niu FN, Zhang X, Li EG, Xu Y, 2010b. Human urinary kallidinogenase suppresses cerebral inflammation in experimental stroke and downregulates nuclear factor-kappaB. J Cereb Blood Flow Metab 30, 1356–1365. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cheng T, Petraglia AL, Li Z, Thiyagarajan M, Zhong Z, Wu Z, Liu D, Maggirwar SB, Deane R, Fernandez JA, LaRue B, Griffin JH, Chopp M, Zlokovic BV, 2006. Activated protein C inhibits tissue plasminogen activator-induced brain hemorrhage. Nat Med 12, 1278–1285. [Abstract] [Google Scholar]
  • Cho JY, Cho SI, Schroff R, Abbott R, Noh JS, Yoon J, Gwag BJ, 2009. A phase I, doubleblinded, randomized, placebo-controlled ascending IV single-dose study assessing the safety, tolerance, and pharmacokinetics of Neu2000 in healthy volunteer. 34th Int Stroke Conference, San Diego Abst P421. [Google Scholar]
  • Cho SI, Park UJ, Chung JM, Gwag BJ, 2010. Neu2000, an NR2B-selective, moderate NMDA receptor antagonist and potent spin trapping molecule for stroke. Drug News Perspect 23, 549–556. [Abstract] [Google Scholar]
  • Choi JH, Bateman BT, Mangla S, Marshall RS, Prabhakaran S, Chong J, Mohr JP, Mast H, Pile-Spellman J, 2006. Endovascular recanalization therapy in acute ischemic stroke. Stroke 37, 419–424. [Abstract] [Google Scholar]
  • Clark WM, Rinker LG, Lessov NS, Hazel K, Hill JK, Stenzel-Poore M, Eckenstein F, 2000. Lack of interleukin-6 expression is not protective against focal central nervous system ischemia. Stroke 31, 1715–1720. [Abstract] [Google Scholar]
  • Cohen JE, Itshayek E, Moskovici S, Gomori JM, Fraifeld S, Eichel R, Leker RR, 2011. State-of-the-art reperfusion strategies for acute ischemic stroke. J Clin Neurosci 18, 319–323. [Abstract] [Google Scholar]
  • Colbourne F, Li H, Buchan AM, Clemens JA, 1999. Continuing postischemic neuronal death in CA1: influence of ischemia duration and cytoprotective doses of NBQX and SNX-111 in rats. Stroke 30, 662–668. [Abstract] [Google Scholar]
  • Collaborators, G.B.D.L.R.o.S., Feigin VL, Nguyen G, Cercy K, Johnson CO, Alam T, Parmar PG, Abajobir AA, Abate KH, Abd-Allah F, Abejie AN, Abyu GY, Ademi Z, Agarwal G, Ahmed MB, Akinyemi RO, Al-Raddadi R, Aminde LN, Amlie-Lefond C, Ansari H, Asayesh H, Asgedom SW, Atey TM, Ayele HT, Banach M, Banerjee A, Barac A, Barker-Collo SL, Barnighausen T, Barregard L, Basu S, Bedi N, Behzadifar M, Bejot Y, Bennett DA, Bensenor IM, Berhe DF, Boneya DJ, Brainin M, Campos-Nonato IR, Caso V, Castaneda-Orjuela CA, Rivas JC, Catala-Lopez F, Christensen H, Criqui MH, Damasceno A, Dandona L, Dandona R, Davletov K, de Courten B, deVeber G, Dokova K, Edessa D, Endres M, Faraon EJA, Farvid MS, Fischer F, Foreman K, Forouzanfar MH, Gall SL, Gebrehiwot TT, Geleijnse JM, Gillum RF, Giroud M, Goulart AC, Gupta R, Gupta R, Hachinski V, Hamadeh RR, Hankey GJ, Hareri HA, Havmoeller R, Hay SI, Hegazy MI, Hibstu DT, James SL, Jeemon P, John D, Jonas JB, Jozwiak J, Kalani R, Kandel A, Kasaeian A, Kengne AP, Khader YS, Khan AR, Khang YH, Khubchandani J, Kim D, Kim YJ, Kivimaki M, Kokubo Y, Kolte D, Kopec JA, Kosen S, Kravchenko M, Krishnamurthi R, Kumar GA, Lafranconi A, Lavados PM, Legesse Y, Li Y, Liang X, Lo WD, Lorkowski S, Lotufo PA, Loy CT, Mackay MT, Abd El Razek HM, Mahdavi M, Majeed A, Malekzadeh R, Malta DC, Mamun AA, Mantovani LG, Martins SCO, Mate KK, Mazidi M, Mehata S, Meier T, Melaku YA, Mendoza W, Mensah GA, Meretoja A, Mezgebe HB, Miazgowski T, Miller TR, Ibrahim NM, Mohammed S, Mokdad AH, Moosazadeh M, Moran AE, Musa KI, Negoi RI, Nguyen M, Nguyen QL, Nguyen TH, Tran TT, Nguyen TT, Anggraini Ningrum DN, Norrving B, Noubiap JJ, O’Donnell MJ, Olagunju AT, Onuma OK, Owolabi MO, Parsaeian M, Patton GC, Piradov M, Pletcher MA, Pourmalek F, Prakash V, Qorbani M, Rahman M, Rahman MA, Rai RK, Ranta A, Rawaf D, Rawaf S, Renzaho AM, Robinson SR, Sahathevan R, Sahebkar A, Salomon JA, Santalucia P, Santos IS, Sartorius B, Schutte AE, Sepanlou SG, Shafieesabet A, Shaikh MA, Shamsizadeh M, Sheth KN, Sisay M, Shin MJ, Shiue I, Silva DAS, Sobngwi E, Soljak M, Sorensen RJD, Sposato LA, Stranges S, Suliankatchi RA, TabaresSeisdedos R, Tanne D, Nguyen CT, Thakur JS, Thrift AG, Tirschwell DL, Topor-Madry R, Tran BX, Nguyen LT, Truelsen T, Tsilimparis N, Tyrovolas S, Ukwaja KN, Uthman OA, Varakin Y, Vasankari T, Venketasubramanian N, Vlassov VV, Wang W, Werdecker A, Wolfe CDA, Xu G, Yano Y, Yonemoto N, Yu C, Zaidi Z, El Sayed Zaki M, Zhou M, Ziaeian B, Zipkin B, Vos T, Naghavi M, Murray CJL, Roth GA, 2018. Global, Regional, and Country-Specific Lifetime Risks of Stroke, 1990 and 2016. N Engl J Med 379, 2429–2437. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cook DJ, Teves L, Tymianski M, 2012. Treatment of stroke with a PSD-95 inhibitor in the gyrencephalic primate brain. Nature 483, 213–217. [Abstract] [Google Scholar]
  • Cui LY, Zhu YC, Gao S, Wang JM, Peng B, Ni J, Zhou LX, He J, Ma XQ, 2013. Ninety-day administration of dl-3-n-butylphthalide for acute ischemic stroke: a randomized, doubleblind trial. Chin Med J (Engl) 126, 3405–3410. [Abstract] [Google Scholar]
  • Cui W, Matsuno K, Iwata K, Ibi M, Katsuyama M, Kakehi T, Sasaki M, Ikami K, Zhu K, Yabe-Nishimura C, 2009. NADPH oxidase isoforms and anti-hypertensive effects of atorvastatin demonstrated in two animal models. J Pharmacol Sci 111, 260–268. [Abstract] [Google Scholar]
  • Czech B, Pfeilschifter W, Mazaheri-Omrani N, Strobel MA, Kahles T, Neumann-Haefelin T, Rami A, Huwiler A, Pfeilschifter J, 2009. The immunomodulatory sphingosine 1-phosphate analog FTY720 reduces lesion size and improves neurological outcome in a mouse model of cerebral ischemia. Biochem Biophys Res Commun 389, 251–256. [Abstract] [Google Scholar]
  • Darsalia V, Mansouri S, Ortsater H, Olverling A, Nozadze N, Kappe C, Iverfeldt K, Tracy LM, Grankvist N, Sjoholm A, Patrone C, 2012. Glucagon-like peptide-1 receptor activation reduces ischaemic brain damage following stroke in Type 2 diabetic rats. Clin Sci (Lond) 122, 473–483. [Europe PMC free article] [Abstract] [Google Scholar]
  • Deb I, Manhas N, Poddar R, Rajagopal S, Allan AM, Lombroso PJ, Rosenberg GA, Candelario-Jalil E, Paul S, 2013. Neuroprotective role of a brain-enriched tyrosine phosphatase, STEP, in focal cerebral ischemia. J Neurosci 33, 17814–17826. [Europe PMC free article] [Abstract] [Google Scholar]
  • Deb I, Poddar R, Paul S, 2011. Oxidative stress-induced oligomerization inhibits the activity of the non-receptor tyrosine phosphatase STEP61. J Neurochem 116, 1097–1111. [Europe PMC free article] [Abstract] [Google Scholar]
  • Del Zoppo GJ, Saver JL, Jauch EC, Adams HP Jr., American Heart Association Stroke, C., 2009. Expansion of the time window for treatment of acute ischemic stroke with intravenous tissue plasminogen activator: a science advisory from the American Heart Association/American Stroke Association. Stroke 40, 2945–2948. [Europe PMC free article] [Abstract] [Google Scholar]
  • Deshmukh R, Sharma V, Mehan S, Sharma N, Bedi KL, 2009. Amelioration of intracerebroventricular streptozotocin induced cognitive dysfunction and oxidative stress by vinpocetine -- a PDE1 inhibitor. Eur J Pharmacol 620, 49–56. [Abstract] [Google Scholar]
  • Dhanesha N, Vazquez-Rosa E, Cintron-Perez CJ, Thedens D, Kort AJ, Chuong V, RiveraDompenciel AM, Chauhan AK, Leira EC, Pieper AA, 2018. Treatment with Uric Acid Reduces Infarct and Improves Neurologic Function in Female Mice After Transient Cerebral Ischemia. J Stroke Cerebrovasc Dis 27, 1412–1416. [Abstract] [Google Scholar]
  • Di X, Huang W, 1998. Localization and morphometric study on motoneuronotrophic factor 1 and its receptor in developing chorionic villi of human placenta. Acta Anatomica Sinica 29, 86–89. [Google Scholar]
  • Di X, Huang W, Sun L, 1997. Immunohistochemical localization of c-fos p53 protein & MNTF1 receptor in early human placental villi. Acta Anatomica Sinica 28, 404–406. [Google Scholar]
  • DiNapoli VA, Huber JD, Houser K, Li X, Rosen CL, 2008. Early disruptions of the bloodbrain barrier may contribute to exacerbated neuronal damage and prolonged functional recovery following stroke in aged rats. Neurobiol Aging 29, 753–764. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ding F, Lai J, Gao Y, Wang G, Shang J, Zhang D, Zheng S, 2019. NEAT1/miR-23a-3p/KLF3: a novel regulatory axis in melanoma cancer progression. Cancer Cell Int 19, 217. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dirnagl U, Fisher M, 2012. REPRINT: International, multicenter randomized preclinical trials in translational stroke research: it is time to act. Stroke 43, 1453–1454. [Abstract] [Google Scholar]
  • Dirnagl U, Iadecola C, Moskowitz MA, 1999. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci 22, 391–397. [Abstract] [Google Scholar]
  • Doeppner TR, Zechmeister B, Kaltwasser B, Jin F, Zheng X, Majid A, Venkataramani V, Bahr M, Hermann DM, 2018. Very Delayed Remote Ischemic Post-conditioning Induces Sustained Neurological Recovery by Mechanisms Involving Enhanced Angioneurogenesis and Peripheral Immunosuppression Reversal. Front Cell Neurosci 12, 383. [Europe PMC free article] [Abstract] [Google Scholar]
  • Donawho CK, Luo Y, Luo Y, Penning TD, Bauch JL, Bouska JJ, Bontcheva-Diaz VD, Cox BF, DeWeese TL, Dillehay LE, Ferguson DC, Ghoreishi-Haack NS, Grimm DR, Guan R, Han EK, Holley-Shanks RR, Hristov B, Idler KB, Jarvis K, Johnson EF, Kleinberg LR, Klinghofer V, Lasko LM, Liu X, Marsh KC, McGonigal TP, Meulbroek JA, Olson AM, Palma JP, Rodriguez LE, Shi Y, Stavropoulos JA, Tsurutani AC, Zhu GD, Rosenberg SH, Giranda VL, Frost DJ, 2007. ABT-888, an orally active poly(ADPribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res 13, 2728–2737. [Abstract] [Google Scholar]
  • Dong M, Yan BP, Liao JK, Lam YY, Yip GW, Yu CM, 2010. Rho-kinase inhibition: a novel therapeutic target for the treatment of cardiovascular diseases. Drug Discov Today 15, 622–629. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dong TF, Lv HX, Niu XL, Gui YK, Zhang P, Yan HQ, Li T, 2016. Effect of Urinary Kallidinogenase on Transforming Growth Factor-beta1 and High-Sensitivity C-Reactive Protein Expression in Rat Focal Cerebral Ischemic Injury. Med Sci Monit 22, 2852–2858. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dong Y, Li H, Dong Q, 2020a. The effect of intravenous ginkgolide on clinical improvement of patients with acute ischemic stroke. Neurol Res 42, 260–266. [Abstract] [Google Scholar]
  • Dong Y, Qu J, Zhang Z, Wang C, Dong Q, 2020b. Human urinary kallidinogenase in treating acute ischemic stroke patients: analyses of pooled data from a randomized double-blind placebocontrolled phase IIb and phase III clinical trial. Neurol Res 42, 286–290. [Abstract] [Google Scholar]
  • Donkor ES, 2018. Stroke in the 21(st) Century: A Snapshot of the Burden, Epidemiology, and Quality of Life. Stroke Res Treat 2018, 3238165. [Europe PMC free article] [Abstract] [Google Scholar]
  • Donnan GA, Fisher M, Macleod M, Davis SM, 2008. Stroke. Lancet 371, 1612–1623. [Abstract] [Google Scholar]
  • Dorans KS, Mills KT, Liu Y, He J, 2018. Trends in Prevalence and Control of Hypertension According to the 2017 American College of Cardiology/American Heart Association (ACC/AHA) Guideline. J Am Heart Assoc 7. [Europe PMC free article] [Abstract] [Google Scholar]
  • Drucker DJ, Nauck MA, 2006. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368, 1696–1705. [Abstract] [Google Scholar]
  • Edaravone Acute Infarction Study, G., 2003. Effect of a novel free radical scavenger, edaravone (MCI186), on acute brain infarction. Randomized, placebo-controlled, double-blind study at multicenters. Cerebrovasc Dis 15, 222–229. [Abstract] [Google Scholar]
  • Eliasson MJ, Sampei K, Mandir AS, Hurn PD, Traystman RJ, Bao J, Pieper A, Wang ZQ, Dawson TM, Snyder SH, Dawson VL, 1997. Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med 3, 1089–1095. [Abstract] [Google Scholar]
  • Elkind MS, 2006. Statins as acute-stroke treatment. Int J Stroke 1, 224–225. [Abstract] [Google Scholar]
  • Elkind MS, Sacco RL, MacArthur RB, Fink DJ, Peerschke E, Andrews H, Neils G, Stillman J, Corporan T, Leifer D, Cheung K, 2008. The Neuroprotection with Statin Therapy for Acute Recovery Trial (NeuSTART): an adaptive design phase I dose-escalation study of highdose lovastatin in acute ischemic stroke. Int J Stroke 3, 210–218. [Europe PMC free article] [Abstract] [Google Scholar]
  • Elkind MS, Sacco RL, Macarthur RB, Peerschke E, Neils G, Andrews H, Stillman J, Corporan T, Leifer D, Liu R, Cheung K, 2009. High-dose lovastatin for acute ischemic stroke: results of the phase I dose escalation neuroprotection with statin therapy for acute recovery trial (NeuSTART). Cerebrovasc Dis 28, 266–275. [Europe PMC free article] [Abstract] [Google Scholar]
  • Elkins J, Veltkamp R, Montaner J, Johnston SC, Singhal AB, Becker K, Lansberg MG, Tang W, Chang I, Muralidharan K, Gheuens S, Mehta L, Elkind MSV, 2017. Safety and efficacy of natalizumab in patients with acute ischaemic stroke (ACTION): a randomised, placebocontrolled, double-blind phase 2 trial. Lancet Neurol 16, 217–226. [Abstract] [Google Scholar]
  • Ellery SJ, Goss MG, Brew N, Dickinson H, Hale N, LaRosa DA, Walker DW, Wong FY, 2019. Evaluation of 3K3A-Activated Protein C to Treat Neonatal Hypoxic Ischemic Brain Injury in the Spiny Mouse. Neurotherapeutics 16, 231–243. [Europe PMC free article] [Abstract] [Google Scholar]
  • Emanuelia C, Madeddu P, 2003. Human tissue kallikrein: a new bullet for the treatment of ischemia. Curr Pharm Des 9, 589–597. [Abstract] [Google Scholar]
  • Endres M, Laufs U, Huang Z, Nakamura T, Huang P, Moskowitz MA, Liao JK, 1998. Stroke protection by 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors mediated by endothelial nitric oxide synthase. Proc Natl Acad Sci U S A 95, 8880–8885. [Europe PMC free article] [Abstract] [Google Scholar]
  • England TJ, Hedstrom A, O’Sullivan S, Donnelly R, Barrett DA, Sarmad S, Sprigg N, Bath PM, 2017. RECAST (Remote Ischemic Conditioning After Stroke Trial): A Pilot Randomized Placebo Controlled Phase II Trial in Acute Ischemic Stroke. Stroke 48, 1412–1415. [Abstract] [Google Scholar]
  • Enomoto M, Endo A, Yatsushige H, Fushimi K, Otomo Y, 2019. Clinical Effects of Early Edaravone Use in Acute Ischemic Stroke Patients Treated by Endovascular Reperfusion Therapy. Stroke 50, 652–658. [Abstract] [Google Scholar]
  • Erdo SL, Cai NS, Wolff JR, Kiss B, 1990. Vinpocetin protects against excitotoxic cell death in primary cultures of rat cerebral cortex. Eur J Pharmacol 187, 551–553. [Abstract] [Google Scholar]
  • Fang W, Sha L, Kodithuwakku ND, Wei J, Zhang R, Han D, Mao L, Li Y, 2015. Attenuated Blood-Brain Barrier Dysfunction by XQ-1H Following Ischemic Stroke in Hyperlipidemic Rats. Mol Neurobiol 52, 162–175. [Abstract] [Google Scholar]
  • Feigin VL, Doronin BM, Popova TF, Gribatcheva EV, Tchervov DV, 2001. Vinpocetine treatment in acute ischaemic stroke: a pilot single-blind randomized clinical trial. Eur J Neurol 8, 81–85. [Abstract] [Google Scholar]
  • Feldman JM, 1985. Review of glyburide after one year on the market. Am J Med 79, 102–108. [Abstract] [Google Scholar]
  • Feng Z, Sun Q, Chen W, Bai Y, Hu D, Xie X, 2019. The neuroprotective mechanisms of ginkgolides and bilobalide in cerebral ischemic injury: a literature review. Mol Med 25, 57. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fischer U, Arnold M, Nedeltchev K, Schoenenberger RA, Kappeler L, Hollinger P, Schroth G, Ballinari P, Mattle HP, 2006. Impact of comorbidity on ischemic stroke outcome. Acta Neurol Scand 113, 108–113. [Abstract] [Google Scholar]
  • Fisher M, Albers GW, Donnan GA, Furlan AJ, Grotta JC, Kidwell CS, Sacco RL, Wechsler LR, Stroke Therapy Academic Industry Roundtable, I.V., 2005. Enhancing the development and approval of acute stroke therapies: Stroke Therapy Academic Industry roundtable. Stroke 36, 1808–1813. [Abstract] [Google Scholar]
  • Forouzanfar MH, Liu P, Roth GA, Ng M, Biryukov S, Marczak L, Alexander L, Estep K, Hassen Abate K, Akinyemiju TF, Ali R, Alvis-Guzman N, Azzopardi P, Banerjee A, Barnighausen T, Basu A, Bekele T, Bennett DA, Biadgilign S, Catala-Lopez F, Feigin VL, Fernandes JC, Fischer F, Gebru AA, Gona P, Gupta R, Hankey GJ, Jonas JB, Judd SE, Khang YH, Khosravi A, Kim YJ, Kimokoti RW, Kokubo Y, Kolte D, Lopez A, Lotufo PA, Malekzadeh R, Melaku YA, Mensah GA, Misganaw A, Mokdad AH, Moran AE, Nawaz H, Neal B, Ngalesoni FN, Ohkubo T, Pourmalek F, Rafay A, Rai RK, Rojas-Rueda D, Sampson UK, Santos IS, Sawhney M, Schutte AE, Sepanlou SG, Shifa GT, Shiue I, Tedla BA, Thrift AG, Tonelli M, Truelsen T, Tsilimparis N, Ukwaja KN, Uthman OA, Vasankari T, Venketasubramanian N, Vlassov VV, Vos T, Westerman R, Yan LL, Yano Y, Yonemoto N, Zaki ME, Murray CJ, 2017. Global Burden of Hypertension and Systolic Blood Pressure of at Least 110 to 115 mm Hg, 1990–2015. JAMA 317, 165–182. [Abstract] [Google Scholar]
  • Fraser JF, Maniskas M, Trout A, Lukins D, Parker L, Stafford WL, Alhajeri A, Roberts J, Bix GJ, 2017. Intra-arterial verapamil post-thrombectomy is feasible, safe, and neuroprotective in stroke. J Cereb Blood Flow Metab 37, 3531–3543. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fredriksson L, Li H, Fieber C, Li X, Eriksson U, 2004. Tissue plasminogen activator is a potent activator of PDGF-CC. EMBO J 23, 3793–3802. [Europe PMC free article] [Abstract] [Google Scholar]
  • Frishman WH, Charlap S, 1983. Verapamil in treatment of chronic stable angina. Arch Intern Med 143, 1407–1415. [Abstract] [Google Scholar]
  • Fu Y, Zhang N, Ren L, Yan Y, Sun N, Li YJ, Han W, Xue R, Liu Q, Hao J, Yu C, Shi FD, 2014. Impact of an immune modulator fingolimod on acute ischemic stroke. Proc Natl Acad Sci U S A 111, 18315–18320. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fukuta T, Asai T, Sato A, Namba M, Yanagida Y, Kikuchi T, Koide H, Shimizu K, Oku N, 2016. Neuroprotection against cerebral ischemia/reperfusion injury by intravenous administration of liposomal fasudil. Int J Pharm 506, 129–137. [Abstract] [Google Scholar]
  • Fukuta T, Asai T, Yanagida Y, Namba M, Koide H, Shimizu K, Oku N, 2017. Combination therapy with liposomal neuroprotectants and tissue plasminogen activator for treatment of ischemic stroke. FASEB J 31, 1879–1890. [Abstract] [Google Scholar]
  • Gagalo I, Rusiecka I, Kocic I, 2015. Tyrosine Kinase Inhibitor as a new Therapy for Ischemic Stroke and other Neurologic Diseases: is there any Hope for a Better Outcome? Curr Neuropharmacol 13, 836–844. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gallacher KI, Batty GD, McLean G, Mercer SW, Guthrie B, May CR, Langhorne P, Mair FS, 2014. Stroke, multimorbidity and polypharmacy in a nationally representative sample of 1,424,378 patients in Scotland: implications for treatment burden. BMC Med 12, 151. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gao S, McMillan RP, Zhu Q, Lopaschuk GD, Hulver MW, Butler AA, 2015. Therapeutic effects of adropin on glucose tolerance and substrate utilization in diet-induced obese mice with insulin resistance. Mol Metab 4, 310–324. [Europe PMC free article] [Abstract] [Google Scholar]
  • Garcia-Bonilla L, Campos M, Giralt D, Salat D, Chacon P, Hernandez-Guillamon M, Rosell A, Montaner J, 2012. Evidence for the efficacy of statins in animal stroke models: a metaanalysis. J Neurochem 122, 233–243. [Abstract] [Google Scholar]
  • Gertz K, Kronenberg G, Kalin RE, Baldinger T, Werner C, Balkaya M, Eom GD, HellmannRegen J, Krober J, Miller KR, Lindauer U, Laufs U, Dirnagl U, Heppner FL, Endres M, 2012. Essential role of interleukin-6 in post-stroke angiogenesis. Brain 135, 1964–1980. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gertz K, Laufs U, Lindauer U, Nickenig G, Bohm M, Dirnagl U, Endres M, 2003. Withdrawal of statin treatment abrogates stroke protection in mice. Stroke 34, 551–557. [Abstract] [Google Scholar]
  • Ginsberg MD, 2008. Neuroprotection for ischemic stroke: past, present and future. Neuropharmacology 55, 363–389. [Europe PMC free article] [Abstract] [Google Scholar]
  • Goke R, Larsen PJ, Mikkelsen JD, Sheikh SP, 1995. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci 7, 2294–2300. [Abstract] [Google Scholar]
  • Griffin JH, Zlokovic BV, Mosnier LO, 2018. Activated protein C, protease activated receptor 1, and neuroprotection. Blood 132, 159–169. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gulke E, Gelderblom M, Magnus T, 2018. Danger signals in stroke and their role on microglia activation after ischemia. Ther Adv Neurol Disord 11, 1756286418774254. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gulyas B, Halldin C, Sandell J, Karlsson P, Sovago J, Karpati E, Kiss B, Vas A, Cselenyi Z, Farde L, 2002a. PET studies on the brain uptake and regional distribution of [11C]vinpocetine in human subjects. Acta Neurol Scand 106, 325–332. [Abstract] [Google Scholar]
  • Gulyas B, Halldin C, Sovago J, Sandell J, Cselenyi Z, Vas A, Kiss B, Karpati E, Farde L, 2002b. Drug distribution in man: a positron emission tomography study after oral administration of the labelled neuroprotective drug vinpocetine. Eur J Nucl Med Mol Imaging 29, 1031–1038. [Abstract] [Google Scholar]
  • Guo H, Liu D, Gelbard H, Cheng T, Insalaco R, Fernandez JA, Griffin JH, Zlokovic BV, 2004. Activated protein C prevents neuronal apoptosis via protease activated receptors 1 and 3. Neuron 41, 563–572. [Abstract] [Google Scholar]
  • Guo H, Singh I, Wang Y, Deane R, Barrett T, Fernandez JA, Chow N, Griffin JH, Zlokovic BV, 2009. Neuroprotective activities of activated protein C mutant with reduced anticoagulant activity. Eur J Neurosci 29, 1119–1130. [Europe PMC free article] [Abstract] [Google Scholar]
  • Guo Z, Yu S, Chen X, Ye R, Zhu W, Liu X, 2016. NLRP3 Is Involved in Ischemia/Reperfusion Injury. CNS Neurol Disord Drug Targets 15, 699–712. [Abstract] [Google Scholar]
  • Guo Z, Yu S, Chen X, Zheng P, Hu T, Duan Z, Liu X, Liu Q, Ye R, Zhu W, Liu X, 2018. Suppression of NLRP3 attenuates hemorrhagic transformation after delayed rtPA treatment in thromboembolic stroke rats: Involvement of neutrophil recruitment. Brain Res Bull 137, 229–240. [Abstract] [Google Scholar]
  • Gursoy-Ozdemir Y, Can A, Dalkara T, 2004. Reperfusion-induced oxidative/nitrative injury to neurovascular unit after focal cerebral ischemia. Stroke 35, 1449–1453. [Abstract] [Google Scholar]
  • Guthrie B, Payne K, Alderson P, McMurdo ME, Mercer SW, 2012. Adapting clinical guidelines to take account of multimorbidity. BMJ 345, e6341. [Abstract] [Google Scholar]
  • Gwag BJ, Lee YA, Ko SY, Lee MJ, Im DS, Yun BS, Lim HR, Park SM, Byun HY, Son SJ, Kwon HJ, Lee JY, Cho JY, Won SJ, Kim KW, Ahn YM, Moon HS, Lee HU, Yoon SH, Noh JH, Chung JM, Cho SI, 2007. Marked prevention of ischemic brain injury by Neu2000, an NMDA antagonist and antioxidant derived from aspirin and sulfasalazine. J Cereb Blood Flow Metab 27, 1142–1151. [Abstract] [Google Scholar]
  • Hagberg H, Wilson MA, Matsushita H, Zhu C, Lange M, Gustavsson M, Poitras MF, Dawson TM, Dawson VL, Northington F, Johnston MV, 2004. PARP-1 gene disruption in mice preferentially protects males from perinatal brain injury. J Neurochem 90, 1068–1075. [Abstract] [Google Scholar]
  • Hamilton A, Holscher C, 2009. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport 20, 1161–1166. [Abstract] [Google Scholar]
  • Hamzei Taj S, Kho W, Riou A, Wiedermann D, Hoehn M, 2016. MiRNA-124 induces neuroprotection and functional improvement after focal cerebral ischemia. Biomaterials 91, 151–165. [Abstract] [Google Scholar]
  • Han L, Li J, Chen Y, Zhang M, Qian L, Chen Y, Wu Z, Xu Y, Li J, 2015. Human Urinary Kallidinogenase Promotes Angiogenesis and Cerebral Perfusion in Experimental Stroke. PLoS One 10, e0134543. [Europe PMC free article] [Abstract] [Google Scholar]
  • Han S, Kim Y, Choi H, Soh D, Kim J, Nam J, Kim J, Bae K, Lim H, 2017. Safety, Tolerability, and Pharmacokinetics of Single and Multiple Dose JPI-289, A Novel Poly (ADPRibose) Polymerase-1 Inhibitor in First-In-Human Study of Healthy Volunteers. Clinical Therapeutics 39, e48. [Google Scholar]
  • Harraz MM, Eacker SM, Wang X, Dawson TM, Dawson VL, 2012. MicroRNA-223 is neuroprotective by targeting glutamate receptors. Proc Natl Acad Sci U S A 109, 18962–18967. [Europe PMC free article] [Abstract] [Google Scholar]
  • Heidenreich PA, Trogdon JG, Khavjou OA, Butler J, Dracup K, Ezekowitz MD, Finkelstein EA, Hong Y, Johnston SC, Khera A, Lloyd-Jones DM, Nelson SA, Nichol G, Orenstein D, Wilson PW, Woo YJ, American Heart Association Advocacy Coordinating, C., Stroke C, Council on Cardiovascular, R., Intervention, Council on Clinical, C., Council on, E., Prevention, Council on, A., Thrombosis, Vascular, B., Council on, C., Critical, C., Perioperative, Resuscitation, Council on Cardiovascular, N., Council on the Kidney in Cardiovascular, D., Council on Cardiovascular, S., Anesthesia, Interdisciplinary Council on Quality of, C., Outcomes, R., 2011. Forecasting the future of cardiovascular disease in the United States: a policy statement from the American Heart Association. Circulation 123, 933–944. [Abstract] [Google Scholar]
  • Helton R, Cui J, Scheel JR, Ellison JA, Ames C, Gibson C, Blouw B, Ouyang L, Dragatsis I, Zeitlin S, Johnson RS, Lipton SA, Barlow C, 2005. Brain-specific knock-out of hypoxiainducible factor-1alpha reduces rather than increases hypoxic-ischemic damage. J Neurosci 25, 4099–4107. [Europe PMC free article] [Abstract] [Google Scholar]
  • Herrmann O, Tarabin V, Suzuki S, Attigah N, Coserea I, Schneider A, Vogel J, Prinz S, Schwab S, Monyer H, Brombacher F, Schwaninger M, 2003. Regulation of body temperature and neuroprotection by endogenous interleukin-6 in cerebral ischemia. J Cereb Blood Flow Metab 23, 406–415. [Abstract] [Google Scholar]
  • Hess DC, Blauenfeldt RA, Andersen G, Hougaard KD, Hoda MN, Ding Y, Ji X, 2015. Remote ischaemic conditioning-a new paradigm of self-protection in the brain. Nat Rev Neurol 11, 698–710. [Abstract] [Google Scholar]
  • Hess DC, Wechsler LR, Clark WM, Savitz SI, Ford GA, Chiu D, Yavagal DR, Uchino K, Liebeskind DS, Auchus AP, Sen S, Sila CA, Vest JD, Mays RW, 2017. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol 16, 360–368. [Abstract] [Google Scholar]
  • Higashi Y, 2009. Edaravone for the treatment of acute cerebral infarction: role of endotheliumderived nitric oxide and oxidative stress. Expert Opin Pharmacother 10, 323–331. [Abstract] [Google Scholar]
  • Higashi Y, Jitsuiki D, Chayama K, Yoshizumi M, 2006. Edaravone (3-methyl-1-phenyl-2pyrazolin-5-one), a novel free radical scavenger, for treatment of cardiovascular diseases. Recent Pat Cardiovasc Drug Discov 1, 85–93. [Abstract] [Google Scholar]
  • Hill MD, Goyal M, Menon BK, Nogueira RG, McTaggart RA, Demchuk AM, Poppe AY, Buck BH, Field TS, Dowlatshahi D, van Adel BA, Swartz RH, Shah RA, Sauvageau E, Zerna C, Ospel JM, Joshi M, Almekhlafi MA, Ryckborst KJ, Lowerison MW, Heard K, Garman D, Haussen D, Cutting SM, Coutts SB, Roy D, Rempel JL, Rohr AC, Iancu D, Sahlas DJ, Yu AYX, Devlin TG, Hanel RA, Puetz V, Silver FL, Campbell BCV, Chapot R, Teitelbaum J, Mandzia JL, Kleinig TJ, Turkel-Parrella D, Heck D, Kelly ME, Bharatha A, Bang OY, Jadhav A, Gupta R, Frei DF, Tarpley JW, McDougall CG, Holmin S, Rha JH, Puri AS, Camden MC, Thomalla G, Choe H, Phillips SJ, Schindler JL, Thornton J, Nagel S, Heo JH, Sohn SI, Psychogios MN, Budzik RF, Starkman S, Martin CO, Burns PA, Murphy S, Lopez GA, English J, Tymianski M, Investigators EN, 2020. Efficacy and safety of nerinetide for the treatment of acute ischaemic stroke (ESCAPENA1): a multicentre, double-blind, randomised controlled trial. Lancet 395, 878–887. [Abstract] [Google Scholar]
  • Hishida A, 2007. Clinical analysis of 207 patients who developed renal disorders during or after treatment with edaravone reported during post-marketing surveillance. Clin Exp Nephrol 11, 292296. [Abstract] [Google Scholar]
  • Hishida A, 2009. Determinants for the prognosis of acute renal disorders that developed during or after treatment with edaravone. Clin Exp Nephrol 13, 118–122. [Abstract] [Google Scholar]
  • Hoda MN, Bhatia K, Hafez SS, Johnson MH, Siddiqui S, Ergul A, Zaidi SK, Fagan SC, Hess DC, 2014. Remote ischemic perconditioning is effective after embolic stroke in ovariectomized female mice. Transl Stroke Res 5, 484–490. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hoda MN, Siddiqui S, Herberg S, Periyasamy-Thandavan S, Bhatia K, Hafez SS, Johnson MH, Hill WD, Ergul A, Fagan SC, Hess DC, 2012. Remote ischemic perconditioning is effective alone and in combination with intravenous tissue-type plasminogen activator in murine model of embolic stroke. Stroke 43, 2794–2799. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hong H, Zeng JS, Kreulen DL, Kaufman DI, Chen AF, 2006. Atorvastatin protects against cerebral infarction via inhibition of NADPH oxidase-derived superoxide in ischemic stroke. Am J Physiol Heart Circ Physiol 291, H2210–2215. [Abstract] [Google Scholar]
  • Hong JM, Choi MH, Sohn SI, Hwang YH, Ahn SH, Lee YB, Shin DI, Chamorro A, Choi DW, on the behalf of the, S.i., 2018. Safety and Optimal Neuroprotection of neu2000 in acute Ischemic stroke with reCanalization: study protocol for a randomized, double-blinded, placebo-controlled, phase-II trial. Trials 19, 375. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hougaard KD, Hjort N, Zeidler D, Sorensen L, Norgaard A, Hansen TM, von WeitzelMudersbach P, Simonsen CZ, Damgaard D, Gottrup H, Svendsen K, Rasmussen PV, Ribe LR, Mikkelsen IK, Nagenthiraja K, Cho TH, Redington AN, Botker HE, Ostergaard L, Mouridsen K, Andersen G, 2014. Remote ischemic perconditioning as an adjunct therapy to thrombolysis in patients with acute ischemic stroke: a randomized trial. Stroke 45, 159–167. [Abstract] [Google Scholar]
  • Howard G, Toole JF, Frye-Pierson J, Hinshelwood LC, 1987. Factors influencing the survival of 451 transient ischemic attack patients. Stroke 18, 552–557. [Abstract] [Google Scholar]
  • Hu J, Wen Q, Wu Y, Li B, Gao P, 2014. The effect of butylphthalide on the brain edema, bloodbrain barrier of rats after focal cerebral infarction and the expression of Rho A. Cell Biochem Biophys 69, 363–368. [Abstract] [Google Scholar]
  • Huang M, Qian Y, Guan T, Huang L, Tang X, Li Y, 2012. Different neuroprotective responses of Ginkgolide B and bilobalide, the two Ginkgo components, in ischemic rats with hyperglycemia. Eur J Pharmacol 677, 71–76. [Abstract] [Google Scholar]
  • Hudobenko J, Chauhan A, McCullough LD, 2017. Interleukin-6 receptor inhibition with tocilizumab ameliorates ischemic stroke damage in mice. Stroke 48, TP270. [Google Scholar]
  • Hudobenko J, Chauhan A, McCullough LD, 2019. Amelioration of ischemic stroke damage through inhibition of interleukin-6 signaling with Tocilizumab requires sex specific dosing. Stroke 50, A128. [Google Scholar]
  • Huffman MD, Lloyd-Jones DM, 2017. Global Burden of Raised Blood Pressure: Coming Into Focus. JAMA 317, 142–143. [Abstract] [Google Scholar]
  • Ivic L, Sands TT, Fishkin N, Nakanishi K, Kriegstein AR, Stromgaard K, 2003. Terpene trilactones from Ginkgo biloba are antagonists of cortical glycine and GABA(A) receptors. J Biol Chem 278, 49279–49285. [Abstract] [Google Scholar]
  • Jacobs BP, Browner WS, 2000. Ginkgo biloba: a living fossil. Am J Med 108, 341–342. [Abstract] [Google Scholar]
  • Jacobs SA, Pinxteren J, Roobrouck VD, Luyckx A, van’t Hof W, Deans R, Verfaillie CM, Waer M, Billiau AD, Van Gool SW, 2013. Human multipotent adult progenitor cells are nonimmunogenic and exert potent immunomodulatory effects on alloreactive T-cell responses. Cell Transplant 22, 1915–1928. [Abstract] [Google Scholar]
  • Jellema RK, Ophelders DR, Zwanenburg A, Nikiforou M, Delhaas T, Andriessen P, Mays RW, Deans R, Germeraad WT, Wolfs TG, Kramer BW, 2015. Multipotent adult progenitor cells for hypoxic-ischemic injury in the preterm brain. J Neuroinflammation 12, 241. [Europe PMC free article] [Abstract] [Google Scholar]
  • Jensen AA, Begum N, Vogensen SB, Knapp KM, Gundertofte K, Dzyuba SV, Ishii H, Nakanishi K, Kristiansen U, Stromgaard K, 2007. Probing the pharmacophore of ginkgolides as glycine receptor antagonists. J Med Chem 50, 1610–1617. [Abstract] [Google Scholar]
  • Jeon KI, Xu X, Aizawa T, Lim JH, Jono H, Kwon DS, Abe J, Berk BC, Li JD, Yan C, 2010. Vinpocetine inhibits NF-kappaB-dependent inflammation via an IKK-dependent but PDEindependent mechanism. Proc Natl Acad Sci U S A 107, 9795–9800. [Europe PMC free article] [Abstract] [Google Scholar]
  • Jin J, Kang HM, Jung J, Jeong JW, Park C, 2014. Related expressional change of HIF-1alpha to the neuroprotective activity of exendin-4 in transient global ischemia. Neuroreport 25, 65–70. [Abstract] [Google Scholar]
  • Jin K, Minami M, Xie L, Sun Y, Mao XO, Wang Y, Simon RP, Greenberg DA, 2004. Ischemia-induced neurogenesis is preserved but reduced in the aged rodent brain. Aging Cell 3, 373–377. [Abstract] [Google Scholar]
  • Jincai W, Tingfang D, Yongheng Z, Zhongmin L, Kaihua Z, Xiaohong L, 2014. Effects of vinpocetine and ozagrel on behavioral recovery of rats after global brain ischemia. J Clin Neurosci 21, 661–663. [Abstract] [Google Scholar]
  • Justicia C, Salas-Perdomo A, Perez-de-Puig I, Deddens LH, van Tilborg GAF, Castellvi C, Dijkhuizen RM, Chamorro A, Planas AM, 2017. Uric Acid Is Protective After Cerebral Ischemia/Reperfusion in Hyperglycemic Mice. Transl Stroke Res 8, 294–305. [Abstract] [Google Scholar]
  • Kanellis J, Johnson RJ, 2003. Editorial comment--Elevated uric acid and ischemic stroke: accumulating evidence that it is injurious and not neuroprotective. Stroke 34, 1956–1957. [Abstract] [Google Scholar]
  • Kanninen KM, Pomeshchik Y, Leinonen H, Malm T, Koistinaho J, Levonen AL, 2015. Applications of the Keap1-Nrf2 system for gene and cell therapy. Free Radic Biol Med 88, 350361. [Abstract] [Google Scholar]
  • Karatepe AG, Gunaydin R, Kaya T, Turkmen G, 2008. Comorbidity in patients after stroke: impact on functional outcome. J Rehabil Med 40, 831–835. [Abstract] [Google Scholar]
  • Katan M, Luft A, 2018. Global Burden of Stroke. Semin Neurol 38, 208–211. [Abstract] [Google Scholar]
  • Kawai H, Deguchi S, Deguchi K, Yamashita T, Ohta Y, Shang J, Tian F, Zhang X, Liu N, Liu W, Ikeda Y, Matsuura T, Abe K, 2011. Synergistic benefit of combined amlodipine plus atorvastatin on neuronal damage after stroke in Zucker metabolic rat. Brain Res 1368, 317–323. [Abstract] [Google Scholar]
  • Kawashima S, Yamashita T, Miwa Y, Ozaki M, Namiki M, Hirase T, Inoue N, Hirata K, Yokoyama M, 2003. HMG-CoA reductase inhibitor has protective effects against stroke events in stroke-prone spontaneously hypertensive rats. Stroke 34, 157–163. [Abstract] [Google Scholar]
  • Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J, 2005. Global burden of hypertension: analysis of worldwide data. Lancet 365, 217–223. [Abstract] [Google Scholar]
  • Kellner CP, Awad AJ, Mocco J, 2016. Developing New Stroke Treatments Using Preclinical Randomized Controlled Trials. World Neurosurg 86, 13–14. [Abstract] [Google Scholar]
  • Keuskamp J, Murali R, Chao KH, 2008. High-dose intraarterial verapamil in the treatment of cerebral vasospasm after aneurysmal subarachnoid hemorrhage. J Neurosurg 108, 458–463. [Abstract] [Google Scholar]
  • Khatri R, McKinney AM, Swenson B, Janardhan V, 2012. Blood-brain barrier, reperfusion injury, and hemorrhagic transformation in acute ischemic stroke. Neurology 79, S52–57. [Abstract] [Google Scholar]
  • Khoshnam SE, Winlow W, Farbood Y, Moghaddam HF, Farzaneh M, 2017. Emerging Roles of microRNAs in Ischemic Stroke: As Possible Therapeutic Agents. J Stroke 19, 166–187. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kikuchi K, Kawahara K, Miyagi N, Uchikado H, Kuramoto T, Morimoto Y, Tancharoen S, Miura N, Takenouchi K, Oyama Y, Shrestha B, Matsuda F, Yoshida Y, Arimura S, Mera K, Tada K, Yoshinaga N, Maenosono R, Ohno Y, Hashiguchi T, Maruyama I, Shigemori M, 2010. Edaravone: a new therapeutic approach for the treatment of acute stroke. Med Hypotheses 75, 583–585. [Abstract] [Google Scholar]
  • Kikuchi K, Setoyama K, Kawahara KI, Nagasato T, Terashi T, Ueda K, Nakanishi K, Otsuka S, Miura N, Sameshima H, Hosokawa K, Harada Y, Shrestha B, Yamamoto M, MorimotoYamashita Y, Kikuchi H, Kiyama R, Kamikokuryo C, Tancharoen S, Sakakima H, Morioka M, Tanaka E, Ito T, Maruyama I, 2017. Edaravone, a Synthetic Free Radical Scavenger, Enhances Alteplase-Mediated Thrombolysis. Oxid Med Cell Longev 2017, 6873281. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kikuchi K, Tancharoen S, Matsuda F, Biswas KK, Ito T, Morimoto Y, Oyama Y, Takenouchi K, Miura N, Arimura N, Nawa Y, Meng X, Shrestha B, Arimura S, Iwata M, Mera K, Sameshima H, Ohno Y, Maenosono R, Tajima Y, Uchikado H, Kuramoto T, Nakayama K, Shigemori M, Yoshida Y, Hashiguchi T, Maruyama I, Kawahara K, 2009. Edaravone attenuates cerebral ischemic injury by suppressing aquaporin-4. Biochem Biophys Res Commun 390, 1121–1125. [Abstract] [Google Scholar]
  • Kim Y, Kim YS, Kim HY, Noh MY, Kim JY, Lee YJ, Kim J, Park J, Kim SH, 2018. Early Treatment with Poly(ADP-Ribose) Polymerase-1 Inhibitor (JPI-289) Reduces Infarct Volume and Improves Long-Term Behavior in an Animal Model of Ischemic Stroke. Mol Neurobiol 55, 7153–7163. [Abstract] [Google Scholar]
  • Kim Y, Kim YS, Noh MY, Lee H, Joe B, Kim HY, Kim J, Kim SH, Park J, 2017. Neuroprotective effects of a novel poly (ADP-ribose) polymerase-1 inhibitor, JPI-289, in hypoxic rat cortical neurons. Clin Exp Pharmacol Physiol 44, 671–679. [Abstract] [Google Scholar]
  • Kimberly WT, Bevers MB, von Kummer R, Demchuk AM, Romero JM, Elm JJ, Hinson HE, Molyneaux BJ, Simard JM, Sheth KN, 2018. Effect of IV glyburide on adjudicated edema endpoints in the GAMES-RP Trial. Neurology 91, e2163–e2169. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kitagawa Y, 2006. Edaravone in acute ischemic stroke. Intern Med 45, 225–226. [Abstract] [Google Scholar]
  • Klein U, Lia M, Crespo M, Siegel R, Shen Q, Mo T, Ambesi-Impiombato A, Califano A, Migliazza A, Bhagat G, Dalla-Favera R, 2010. The DLEU2/miR-15a/16–1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia. Cancer Cell 17, 28–40. [Abstract] [Google Scholar]
  • Koch E, 2005. Inhibition of platelet activating factor (PAF)-induced aggregation of human thrombocytes by ginkgolides: considerations on possible bleeding complications after oral intake of Ginkgo biloba extracts. Phytomedicine 12, 10–16. [Abstract] [Google Scholar]
  • Koh SH, Park Y, Song CW, Kim JG, Kim K, Kim J, Kim MH, Lee SR, Kim DW, Yu HJ, Chang DI, Hwang SJ, Kim SH, 2004. The effect of PARP inhibitor on ischaemic cell death, its related inflammation and survival signals. Eur J Neurosci 20, 1461–1472. [Abstract] [Google Scholar]
  • Kraft P, Gob E, Schuhmann MK, Gobel K, Deppermann C, Thielmann I, Herrmann AM, Lorenz K, Brede M, Stoll G, Meuth SG, Nieswandt B, Pfeilschifter W, Kleinschnitz C, 2013. FTY720 ameliorates acute ischemic stroke in mice by reducing thrombo-inflammation but not by direct neuroprotection. Stroke 44, 3202–3210. [Abstract] [Google Scholar]
  • Kramer W, Muller G, Girbig F, Gutjahr U, Kowalewski S, Hartz D, Summ HD, 1995. The molecular interaction of sulfonylureas with beta-cell ATP-sensitive K(+)-channels. Diabetes Res Clin Pract 28 Suppl, S67–80. [Abstract] [Google Scholar]
  • Krupinski J, Slevin M, 2013. Emerging molecular targets for brain repair after stroke. Stroke Res Treat 2013, 473416. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kumar KG, Trevaskis JL, Lam DD, Sutton GM, Koza RA, Chouljenko VN, Kousoulas KG, Rogers PM, Kesterson RA, Thearle M, Ferrante AW Jr., Mynatt RL, Burris TP, Dong JZ, Halem HA, Culler MD, Heisler LK, Stephens JM, Butler AA, 2008. Identification of adropin as a secreted factor linking dietary macronutrient intake with energy homeostasis and lipid metabolism. Cell Metab 8, 468–481. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kurland DB, Tosun C, Pampori A, Karimy JK, Caffes NM, Gerzanich V, Simard JM, 2013. Glibenclamide for the treatment of acute CNS injury. Pharmaceuticals (Basel) 6, 1287–1303. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kuroki T, Tanaka R, Shimada Y, Yamashiro K, Ueno Y, Shimura H, Urabe T, Hattori N, 2016. Exendin-4 Inhibits Matrix Metalloproteinase-9 Activation and Reduces Infarct Growth After Focal Cerebral Ischemia in Hyperglycemic Mice. Stroke 47, 1328–1335. [Abstract] [Google Scholar]
  • Kwiatkowski TG, Libman RB, Frankel M, Tilley BC, Morgenstern LB, Lu M, Broderick JP, Lewandowski CA, Marler JR, Levine SR, Brott T, 1999. Effects of tissue plasminogen activator for acute ischemic stroke at one year. National Institute of Neurological Disorders and Stroke Recombinant Tissue Plasminogen Activator Stroke Study Group. N Engl J Med 340, 17811787. [Abstract] [Google Scholar]
  • Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T, 2002. Identification of tissue-specific microRNAs from mouse. Curr Biol 12, 735–739. [Abstract] [Google Scholar]
  • Lakhan SE, Kirchgessner A, Hofer M, 2009. Inflammatory mechanisms in ischemic stroke: therapeutic approaches. J Transl Med 7, 97. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lan Z, Xu X, Xu W, Li J, Liang Z, Zhang X, Lei M, Zhao C, 2015. Discovery of 3-n-butyl2,3-dihydro-1H-isoindol-1-one as a potential anti-ischemic stroke agent. Drug Des Devel Ther 9, 3377–3391. [Europe PMC free article] [Abstract] [Google Scholar]
  • Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, Lin C, Socci ND, Hermida L, Fulci V, Chiaretti S, Foa R, Schliwka J, Fuchs U, Novosel A, Muller RU, Schermer B, Bissels U, Inman J, Phan Q, Chien M, Weir DB, Choksi R, De Vita G, Frezzetti D, Trompeter HI, Hornung V, Teng G, Hartmann G, Palkovits M, Di Lauro R, Wernet P, Macino G, Rogler CE, Nagle JW, Ju J, Papavasiliou FN, Benzing T, Lichter P, Tam W, Brownstein MJ, Bosio A, Borkhardt A, Russo JJ, Sander C, Zavolan M, Tuschl T, 2007. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414. [Europe PMC free article] [Abstract] [Google Scholar]
  • Langhauser F, Kraft P, Gob E, Leinweber J, Schuhmann MK, Lorenz K, Gelderblom M, Bittner S, Meuth SG, Wiendl H, Magnus T, Kleinschnitz C, 2014. Blocking of alpha4 integrin does not protect from acute ischemic stroke in mice. Stroke 45, 1799–1806. [Abstract] [Google Scholar]
  • Lapchak PA, 2010. A critical assessment of edaravone acute ischemic stroke efficacy trials: is edaravone an effective neuroprotective therapy? Expert Opin Pharmacother 11, 1753–1763. [Europe PMC free article] [Abstract] [Google Scholar]
  • Laufs U, Gertz K, Dirnagl U, Bohm M, Nickenig G, Endres M, 2002. Rosuvastatin, a new HMG-CoA reductase inhibitor, upregulates endothelial nitric oxide synthase and protects from ischemic stroke in mice. Brain Res 942, 23–30. [Abstract] [Google Scholar]
  • Lawes CM, Vander Hoorn S, Rodgers A, International Society of H, 2008. Global burden of blood-pressure-related disease, 2001. Lancet 371, 1513–1518. [Abstract] [Google Scholar]
  • Lee AS, 2005. The ER chaperone and signaling regulator GRP78/BiP as a monitor of endoplasmic reticulum stress. Methods 35, 373–381. [Abstract] [Google Scholar]
  • Lee BJ, Egi Y, van Leyen K, Lo EH, Arai K, 2010. Edaravone, a free radical scavenger, protects components of the neurovascular unit against oxidative stress in vitro. Brain Res 1307, 2227. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lee CH, Yan B, Yoo KY, Choi JH, Kwon SH, Her S, Sohn Y, Hwang IK, Cho JH, Kim YM, Won MH, 2011. Ischemia-induced changes in glucagon-like peptide-1 receptor and neuroprotective effect of its agonist, exendin-4, in experimental transient cerebral ischemia. J Neurosci Res 89, 1103–1113. [Abstract] [Google Scholar]
  • Lee M, Cho S, La S, Gwag BJ, 2007. Marked neuroprotection of Neu2000 against a rat permanent middle cerebral occlusion model with wide therapeutic time window. 37th Annu Meet Soc Neurosci, San Diego Abst 900.1/X1. [Google Scholar]
  • Lee MMY, Sattar N, McMurray JJV, Packard CJ, 2019. Statins in the Prevention and Treatment of Heart Failure: a Review of the Evidence. Curr Atheroscler Rep 21, 41. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li D, Liu X, Lin L, Hou J, Li N, Wang C, Wang P, Zhang Q, Zhang P, Zhou W, Wang Z, Ding G, Zhuang SM, Zheng L, Tao W, Cao X, 2011. MicroRNA-99a inhibits hepatocellular carcinoma growth and correlates with prognosis of patients with hepatocellular carcinoma. J Biol Chem 286, 36677–36685. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li F, Ma Q, Zhao H, Wang R, Tao Z, Fan Z, Zhang S, Li G, Luo Y, 2018. L-3-nButylphthalide reduces ischemic stroke injury and increases M2 microglial polarization. Metab Brain Dis 33, 1995–2003. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li M, Hou W, Zhang X, Hu L, Tang Z, 2014. Hyperuricemia and risk of stroke: a systematic review and meta-analysis of prospective studies. Atherosclerosis 232, 265–270. [Abstract] [Google Scholar]
  • Li P, Shen M, Gao F, Wu J, Zhang J, Teng F, Zhang C, 2017a. An Antagomir to MicroRNA106b-5p Ameliorates Cerebral Ischemia and Reperfusion Injury in Rats Via Inhibiting Apoptosis and Oxidative Stress. Mol Neurobiol 54, 2901–2921. [Abstract] [Google Scholar]
  • Li PC, Liu LF, Jou MJ, Wang HK, 2016. The GLP-1 receptor agonists exendin-4 and liraglutide alleviate oxidative stress and cognitive and micturition deficits induced by middle cerebral artery occlusion in diabetic mice. BMC Neurosci 17, 37. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li R, Huang C, Chen J, Guo Y, Tan S, 2015. The role of uric acid as a potential neuroprotectant in acute ischemic stroke: a review of literature. Neurol Sci 36, 1097–1103. [Abstract] [Google Scholar]
  • Li S, Zhang X, Fang Q, Zhou J, Zhang M, Wang H, Chen Y, Xu B, Wu Y, Qian L, Xu Y, 2017b. Ginkgo biloba extract improved cognitive and neurological functions of acute ischaemic stroke: a randomised controlled trial. Stroke Vasc Neurol 2, 189–197. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, Powers K, Shen H, Egan JM, Sambamurti K, Brossi A, Lahiri DK, Mattson MP, Hoffer BJ, Wang Y, Greig NH, 2009. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A 106, 1285–1290. [Europe PMC free article] [Abstract] [Google Scholar]
  • Liao SJ, Lin JW, Pei Z, Liu CL, Zeng JS, Huang RX, 2009. Enhanced angiogenesis with dl3n-butylphthalide treatment after focal cerebral ischemia in RHRSP. Brain Res 1289, 69–78. [Abstract] [Google Scholar]
  • Liesz A, Sun L, Zhou W, Schwarting S, Mracsko E, Zorn M, Bauer H, Sommer C, Veltkamp R, 2011a. FTY720 reduces post-ischemic brain lymphocyte influx but does not improve outcome in permanent murine cerebral ischemia. PLoS One 6, e21312. [Europe PMC free article] [Abstract] [Google Scholar]
  • Liesz A, Zhou W, Mracsko E, Karcher S, Bauer H, Schwarting S, Sun L, Bruder D, Stegemann S, Cerwenka A, Sommer C, Dalpke AH, Veltkamp R, 2011b. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain 134, 704–720. [Abstract] [Google Scholar]
  • Liu D, Cheng T, Guo H, Fernandez JA, Griffin JH, Song X, Zlokovic BV, 2004. Tissue plasminogen activator neurovascular toxicity is controlled by activated protein C. Nat Med 10, 1379–1383. [Abstract] [Google Scholar]
  • Liu F, Gong J, Huang W, Wang Z, Wang M, Yang J, Wu C, Wu Z, Han B, 2014a. MicroRNA-106b-5p boosts glioma tumorigensis by targeting multiple tumor suppressor genes. Oncogene 33, 4813–4822. [Abstract] [Google Scholar]
  • Liu J, Zhang C, Tao W, Liu M, 2013. Systematic review and meta-analysis of the efficacy of sphingosine-1-phosphate (S1P) receptor agonist FTY720 (fingolimod) in animal models of stroke. Int J Neurosci 123, 163–169. [Abstract] [Google Scholar]
  • Liu L, Liu H, Yang F, Chen G, Zhou H, Tang M, Zhang R, Dong Q, 2011. Tissue kallikrein protects cortical neurons against hypoxia/reoxygenation injury via the ERK1/2 pathway. Biochem Biophys Res Commun 407, 283–287. [Abstract] [Google Scholar]
  • Liu L, Zhang R, Liu K, Zhou H, Yang X, Liu X, Tang M, Su J, Dong Q, 2009. Tissue kallikrein protects cortical neurons against in vitro ischemia-acidosis/reperfusion-induced injury through the ERK1/2 pathway. Exp Neurol 219, 453–465. [Abstract] [Google Scholar]
  • Liu P, Zhao H, Wang R, Wang P, Tao Z, Gao L, Yan F, Liu X, Yu S, Ji X, Luo Y, 2015. MicroRNA-424 protects against focal cerebral ischemia and reperfusion injury in mice by suppressing oxidative stress. Stroke 46, 513–519. [Abstract] [Google Scholar]
  • Liu X, Zhao S, Liu F, Kang J, Xiao A, Li F, Zhang C, Yan F, Zhao H, Luo M, Luo Y, Ji X, 2014b. Remote ischemic postconditioning alleviates cerebral ischemic injury by attenuating endoplasmic reticulum stress-mediated apoptosis. Transl Stroke Res 5, 692–700. [Abstract] [Google Scholar]
  • Llovera G, Hofmann K, Roth S, Salas-Perdomo A, Ferrer-Ferrer M, Perego C, Zanier ER, Mamrak U, Rex A, Party H, Agin V, Fauchon C, Orset C, Haelewyn B, De Simoni MG, Dirnagl U, Grittner U, Planas AM, Plesnila N, Vivien D, Liesz A, 2015. Results of a preclinical randomized controlled multicenter trial (pRCT): Anti-CD49d treatment for acute brain ischemia. Sci Transl Med 7, 299ra121. [Abstract] [Google Scholar]
  • Lobb RR, Hemler ME, 1994. The pathophysiologic role of alpha 4 integrins in vivo. J Clin Invest 94, 1722–1728. [Europe PMC free article] [Abstract] [Google Scholar]
  • Loddick SA, Turnbull AV, Rothwell NJ, 1998. Cerebral interleukin-6 is neuroprotective during permanent focal cerebral ischemia in the rat. J Cereb Blood Flow Metab 18, 176–179. [Abstract] [Google Scholar]
  • Lombroso PJ, Naegele JR, Sharma E, Lerner M, 1993. A protein tyrosine phosphatase expressed within dopaminoceptive neurons of the basal ganglia and related structures. J Neurosci 13, 3064–3074. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lovren F, Pan Y, Quan A, Singh KK, Shukla PC, Gupta M, Al-Omran M, Teoh H, Verma S, 2010. Adropin is a novel regulator of endothelial function. Circulation 122, S185–192. [Abstract] [Google Scholar]
  • Lovshin JA, Drucker DJ, 2009. Incretin-based therapies for type 2 diabetes mellitus. Nat Rev Endocrinol 5, 262–269. [Abstract] [Google Scholar]
  • Lv P, Fang W, Geng X, Yang Q, Li Y, Sha L, 2011. Therapeutic neuroprotective effects of ginkgolide B on cortex and basal ganglia in a rat model of transient focal ischemia. Eur J Pharm Sci 44, 235–240. [Abstract] [Google Scholar]
  • Lyden P, Levy H, Weymer S, Pryor K, Kramer W, Griffin JH, Davis TP, Zlokovic B, 2013. Phase 1 safety, tolerability and pharmacokinetics of 3K3A-APC in healthy adult volunteers. Curr Pharm Des 19, 7479–7485. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lyden P, Pryor KE, Coffey CS, Cudkowicz M, Conwit R, Jadhav A, Sawyer RN Jr., Claassen J, Adeoye O, Song S, Hannon P, Rost NS, Hinduja A, Torbey M, Lee JM, Benesch C, Rippee M, Rymer M, Froehler MT, Clarke Haley E, Johnson M, Yankey J, Magee K, Qidwai J, Levy H, Mark Haacke E, Fawaz M, Davis TP, Toga AW, Griffin JH, Zlokovic BV, Neuro NCTNNNI, 2019. Final Results of the RHAPSODY Trial: A Multi-Center, Phase 2 Trial Using a Continual Reassessment Method to Determine the Safety and Tolerability of 3K3A-APC, A Recombinant Variant of Human Activated Protein C, in Combination with Tissue Plasminogen Activator, Mechanical Thrombectomy or both in Moderate to Severe Acute Ischemic Stroke. Ann Neurol 85, 125–136. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ma D, Feng L, Deng F, Feng JC, 2017. Overview of Experimental and Clinical Findings regarding the Neuroprotective Effects of Cerebral Ischemic Postconditioning. Biomed Res Int 2017, 6891645. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ma F, Sun P, Zhang X, Hamblin MH, Yin KJ, 2020. Endothelium-targeted deletion of the miR15a/16–1 cluster ameliorates blood-brain barrier dysfunction in ischemic stroke. Sci Signal 13. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ma S, Yin H, Chen L, Liu H, Zhao M, Zhang X, 2012. Neuroprotective effect of ginkgolide K against acute ischemic stroke on middle cerebral ischemia occlusion in rats. J Nat Med 66, 25–31. [Abstract] [Google Scholar]
  • Madden JA, 2012. Role of the vascular endothelium and plaque in acute ischemic stroke. Neurology 79, S58–62. [Abstract] [Google Scholar]
  • Malhotra S, Naggar I, Stewart M, Rosenbaum DM, 2011. Neurogenic pathway mediated remote preconditioning protects the brain from transient focal ischemic injury. Brain Res 1386, 184–190. [Abstract] [Google Scholar]
  • Maniskas ME, Roberts JM, Aron I, Fraser JF, Bix GJ, 2016. Stroke neuroprotection revisited: Intra-arterial verapamil is profoundly neuroprotective in experimental acute ischemic stroke. J Cereb Blood Flow Metab 36, 721–730. [Europe PMC free article] [Abstract] [Google Scholar]
  • Marble A, 1971. Glibenclamide, a new sulphonylurea: whither oral hypoglycaemic agents? Drugs 1, 109–115. [Abstract] [Google Scholar]
  • Marlet IR, Olmestig JNE, Vilsboll T, Rungby J, Kruuse C, 2018. Neuroprotective Mechanisms of Glucagon-like Peptide-1-based Therapies in Ischaemic Stroke: A Systematic Review based on Pre-Clinical Studies. Basic Clin Pharmacol Toxicol 122, 559–569. [Abstract] [Google Scholar]
  • Mayanagi K, Katakam PV, Gaspar T, Domoki F, Busija DW, 2008. Acute treatment with rosuvastatin protects insulin resistant (C57BL/6J ob/ob) mice against transient cerebral ischemia. J Cereb Blood Flow Metab 28, 1927–1935. [Europe PMC free article] [Abstract] [Google Scholar]
  • McCullough LD, Zeng Z, Blizzard KK, Debchoudhury I, Hurn PD, 2005. Ischemic nitric oxide and poly (ADP-ribose) polymerase-1 in cerebral ischemia: male toxicity, female protection. J Cereb Blood Flow Metab 25, 502–512. [Abstract] [Google Scholar]
  • McTavish D, Sorkin EM, 1989. Verapamil. An updated review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in hypertension. Drugs 38, 19–76. [Abstract] [Google Scholar]
  • Medina AE, 2011. Therapeutic utility of phosphodiesterase type I inhibitors in neurological conditions. Front Neurosci 5, 21. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mehta RI, Tosun C, Ivanova S, Tsymbalyuk N, Famakin BM, Kwon MS, Castellani RJ, Gerzanich V, Simard JM, 2015. Sur1-Trpm4 Cation Channel Expression in Human Cerebral Infarcts. J Neuropathol Exp Neurol 74, 835–849. [Europe PMC free article] [Abstract] [Google Scholar]
  • Merali Z, Leung J, Mikulis D, Silver F, Kassner A, 2015. Longitudinal assessment of imatinib’s effect on the blood-brain barrier after ischemia/reperfusion injury with permeability MRI. Transl Stroke Res 6, 39–49. [Abstract] [Google Scholar]
  • Mikeladze KG, Eliava S, Shekhtman OD, Lubnin A, Tabasaranskii TF, Iakovlev SB, 2013. [Intra-arterial injection of verapamil in treatment of cerebral vasospasm in a patient with acute subarachnoid hemorrhage from an aneurysm: case report]. Zh Vopr Neirokhir Im N N Burdenko 77, 57–60; discussion 60. [Abstract] [Google Scholar]
  • Miska EA, Alvarez-Saavedra E, Townsend M, Yoshii A, Sestan N, Rakic P, ConstantinePaton M, Horvitz HR, 2004. Microarray analysis of microRNA expression in the developing mammalian brain. Genome Biol 5, R68. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mizuma A, You JS, Yenari MA, 2018. Targeting Reperfusion Injury in the Age of Mechanical Thrombectomy. Stroke 49, 1796–1802. [Europe PMC free article] [Abstract] [Google Scholar]
  • Molnar P, Erdo SL, 1995. Vinpocetine is as potent as phenytoin to block voltage-gated Na+ channels in rat cortical neurons. Eur J Pharmacol 273, 303–306. [Abstract] [Google Scholar]
  • Moroni F, 2008. Poly(ADP-ribose)polymerase 1 (PARP-1) and postischemic brain damage. Curr Opin Pharmacol 8, 96–103. [Abstract] [Google Scholar]
  • Mosnier LO, Gale AJ, Yegneswaran S, Griffin JH, 2004. Activated protein C variants with normal cytoprotective but reduced anticoagulant activity. Blood 104, 1740–1744. [Abstract] [Google Scholar]
  • Mueller BK, Mack H, Teusch N, 2005. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov 4, 387–398. [Abstract] [Google Scholar]
  • Muller C, Cheung NW, Dewey H, Churilov L, Middleton S, Thijs V, Ekinci EI, Levi C, Lindley R, Donnan G, Parsons M, Bladin C, 2018. Treatment with exenatide in acute ischemic stroke trial protocol: A prospective, randomized, open label, blinded end-point study of exenatide vs. standard care in post stroke hyperglycemia. Int J Stroke 13, 857–862. [Abstract] [Google Scholar]
  • Nada SE, Shah ZA, 2012. Preconditioning with Ginkgo biloba (EGb 761(R)) provides neuroprotection through HO1 and CRMP2. Neurobiol Dis 46, 180–189. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nadel S, Goldstein B, Williams MD, Dalton H, Peters M, Macias WL, Abd-Allah SA, Levy H, Angle R, Wang D, Sundin DP, Giroir B, Sepsis R.E.s., Organ dysfunction in children: a gLobal perspective study, g., 2007. Drotrecogin alfa (activated) in children with severe sepsis: a multicentre phase III randomised controlled trial. Lancet 369, 836–843. [Abstract] [Google Scholar]
  • Nagotani S, Hayashi T, Sato K, Zhang W, Deguchi K, Nagano I, Shoji M, Abe K, 2005. Reduction of cerebral infarction in stroke-prone spontaneously hypertensive rats by statins associated with amelioration of oxidative stress. Stroke 36, 670–672. [Abstract] [Google Scholar]
  • Nakayama H, Jorgensen HS, Raaschou HO, Olsen TS, 1994. The influence of age on stroke outcome. The Copenhagen Stroke Study. Stroke 25, 808–813. [Abstract] [Google Scholar]
  • Nakka VP, Prakash-Babu P, Vemuganti R, 2016. Crosstalk Between Endoplasmic Reticulum Stress, Oxidative Stress, and Autophagy: Potential Therapeutic Targets for Acute CNS Injuries. Mol Neurobiol 53, 532–544. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nazir S, Gadi I, Al-Dabet MM, Elwakiel A, Kohli S, Ghosh S, Manoharan J, Ranjan S, Bock F, Braun-Dullaeus RC, Esmon CT, Huber TB, Camerer E, Dockendorff C, Griffin JH, Isermann B, Shahzad K, 2017. Cytoprotective activated protein C averts Nlrp3 inflammasome-induced ischemia-reperfusion injury via mTORC1 inhibition. Blood 130, 2664–2677. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nelson CW, Wei EP, Povlishock JT, Kontos HA, Moskowitz MA, 1992. Oxygen radicals in cerebral ischemia. Am J Physiol 263, H1356–1362. [Abstract] [Google Scholar]
  • Ng GY, Lim YA, Sobey CG, Dheen T, Fann DY, Arumugam TV, 2018. Epigenetic regulation of inflammation in stroke. Ther Adv Neurol Disord 11, 1756286418771815. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nguyen TH, Liu J, Lombroso PJ, 2002. Striatal enriched phosphatase 61 dephosphorylates Fyn at phosphotyrosine 420. J Biol Chem 277, 24274–24279. [Abstract] [Google Scholar]
  • Ni J, Qu J, Yao M, Zhang Z, Zhong X, Cui L, investigators R, 2017. Re-evaluate the Efficacy and Safety of Human Urinary Kallidinogenase (RESK): Protocol for an Open-Label, Single-Arm, Multicenter Phase IV Trial for the Treatment of Acute Ischemic Stroke in Chinese Patients. Transl Stroke Res 8, 341–346. [Abstract] [Google Scholar]
  • Ni J, Wang X, Chen S, Liu H, Wang Y, Xu X, Cheng J, Jia J, Zhen X, 2015. MicroRNA let-7c-5p protects against cerebral ischemia injury via mechanisms involving the inhibition of microglia activation. Brain Behav Immun 49, 75–85. [Abstract] [Google Scholar]
  • Nishi H, Watanabe T, Sakurai H, Yuki S, Ishibashi A, 1989. Effect of MCI-186 on brain edema in rats. Stroke 20, 1236–1240. [Abstract] [Google Scholar]
  • Noh J, Lee ES, Chung JM, 2009. The novel NMDA receptor antagonist, 2-hydroxy-5-(2,3,5,6tetrafluoro-4-trifluoromethyl-benzylamino)-benzoic acid, is a gating modifier in cultured mouse cortical neurons. J Neurochem 109, 1261–1271. [Abstract] [Google Scholar]
  • Noh MY, Lee WM, Lee SJ, Kim HY, Kim SH, Kim YS, 2018. Regulatory T cells increase after treatment with poly (ADP-ribose) polymerase-1 inhibitor in ischemic stroke patients. Int Immunopharmacol 60, 104–110. [Abstract] [Google Scholar]
  • O’Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW, 2006. 1,026 experimental treatments in acute stroke. Ann Neurol 59, 467–477. [Abstract] [Google Scholar]
  • O’Donnell MJ, Chin SL, Rangarajan S, Xavier D, Liu L, Zhang H, Rao-Melacini P, Zhang X, Pais P, Agapay S, Lopez-Jaramillo P, Damasceno A, Langhorne P, McQueen MJ, Rosengren A, Dehghan M, Hankey GJ, Dans AL, Elsayed A, Avezum A, Mondo C, Diener HC, Ryglewicz D, Czlonkowska A, Pogosova N, Weimar C, Iqbal R, Diaz R, Yusoff K, Yusufali A, Oguz A, Wang X, Penaherrera E, Lanas F, Ogah OS, Ogunniyi A, Iversen HK, Malaga G, Rumboldt Z, Oveisgharan S, Al Hussain F, Magazi D, Nilanont Y, Ferguson J, Pare G, Yusuf S, investigators I, 2016. Global and regional effects of potentially modifiable risk factors associated with acute stroke in 32 countries (INTERSTROKE): a casecontrol study. Lancet 388, 761–775. [Abstract] [Google Scholar]
  • O’Donnell MJ, Xavier D, Liu L, Zhang H, Chin SL, Rao-Melacini P, Rangarajan S, Islam S, Pais P, McQueen MJ, Mondo C, Damasceno A, Lopez-Jaramillo P, Hankey GJ, Dans AL, Yusoff K, Truelsen T, Diener HC, Sacco RL, Ryglewicz D, Czlonkowska A, Weimar C, Wang X, Yusuf S, investigators I, 2010. Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): a case-control study. Lancet 376, 112–123. [Abstract] [Google Scholar]
  • Onetti Y, Dantas AP, Perez B, Cugota R, Chamorro A, Planas AM, Vila E, Jimenez-Altayo F, 2015. Middle cerebral artery remodeling following transient brain ischemia is linked to early postischemic hyperemia: a target of uric acid treatment. Am J Physiol Heart Circ Physiol 308, H862–874. [Abstract] [Google Scholar]
  • Ortega FJ, Gimeno-Bayon J, Espinosa-Parrilla JF, Carrasco JL, Batlle M, Pugliese M, Mahy N, Rodriguez MJ, 2012. ATP-dependent potassium channel blockade strengthens microglial neuroprotection after hypoxia-ischemia in rats. Exp Neurol 235, 282–296. [Abstract] [Google Scholar]
  • Ortega FJ, Jolkkonen J, Mahy N, Rodriguez MJ, 2013. Glibenclamide enhances neurogenesis and improves long-term functional recovery after transient focal cerebral ischemia. J Cereb Blood Flow Metab 33, 356–364. [Europe PMC free article] [Abstract] [Google Scholar]
  • Oskouei DS, Rikhtegar R, Hashemilar M, Sadeghi-Bazargani H, Sharifi-Bonab M, SadeghiHokmabadi E, Zarrintan S, Sharifipour E, 2013. The effect of Ginkgo biloba on functional outcome of patients with acute ischemic stroke: a double-blind, placebo-controlled, randomized clinical trial. J Stroke Cerebrovasc Dis 22, e557–563. [Abstract] [Google Scholar]
  • Ouyang YB, Lu Y, Yue S, Xu LJ, Xiong XX, White RE, Sun X, Giffard RG, 2012. miR181 regulates GRP78 and influences outcome from cerebral ischemia in vitro and in vivo. Neurobiol Dis 45, 555–563. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ouyang YB, Xu L, Lu Y, Sun X, Yue S, Xiong XX, Giffard RG, 2013. Astrocyte-enriched miR-29a targets PUMA and reduces neuronal vulnerability to forebrain ischemia. Glia 61, 1784–1794. [Europe PMC free article] [Abstract] [Google Scholar]
  • Panten U, Schwanstecher M, Schwanstecher C, 1996. Sulfonylurea receptors and mechanism of sulfonylurea action. Exp Clin Endocrinol Diabetes 104, 1–9. [Abstract] [Google Scholar]
  • Pasalic D, Marinkovic N, Feher-Turkovic L, 2012. Uric acid as one of the important factors in multifactorial disorders--facts and controversies. Biochem Med (Zagreb) 22, 63–75. [Europe PMC free article] [Abstract] [Google Scholar]
  • Paterno R, Faraci FM, Heistad DD, 1996. Role of Ca(2+)-dependent K+ channels in cerebral vasodilatation induced by increases in cyclic GMP and cyclic AMP in the rat. Stroke 27, 1603–1607; discussion 1607–1608. [Abstract] [Google Scholar]
  • Paugh SW, Payne SG, Barbour SE, Milstien S, Spiegel S, 2003. The immunosuppressant FTY720 is phosphorylated by sphingosine kinase type 2. FEBS Lett 554, 189–193. [Abstract] [Google Scholar]
  • Paul S, Nairn AC, Wang P, Lombroso PJ, 2003. NMDA-mediated activation of the tyrosine phosphatase STEP regulates the duration of ERK signaling. Nat Neurosci 6, 34–42. [Abstract] [Google Scholar]
  • Paul S, Snyder GL, Yokakura H, Picciotto MR, Nairn AC, Lombroso PJ, 2000. The Dopamine/D1 receptor mediates the phosphorylation and inactivation of the protein tyrosine phosphatase STEP via a PKA-dependent pathway. J Neurosci 20, 5630–5638. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pergakis M, Badjatia N, Chaturvedi S, Cronin CA, Kimberly WT, Sheth KN, Simard JM, 2019. BIIB093 (IV glibenclamide): an investigational compound for the prevention and treatment of severe cerebral edema. Expert Opin Investig Drugs 28, 1031–1040. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pham TH, Okada T, Matloubian M, Lo CG, Cyster JG, 2008. S1P1 receptor signaling overrides retention mediated by G alpha i-coupled receptors to promote T cell egress. Immunity 28, 122–133. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pico F, Lapergue B, Ferrigno M, Rosso C, Meseguer E, Chadenat ML, Bourdain F, Obadia M, Hirel C, Duong DL, Deltour S, Aegerter P, Labreuche J, Cattenoy A, Smadja D, Hosseini H, Guillon B, Wolff V, Samson Y, Cordonnier C, Amarenco P, 2020. Effect of In-Hospital Remote Ischemic Perconditioning on Brain Infarction Growth and Clinical Outcomes in Patients With Acute Ischemic Stroke: The RESCUE BRAIN Randomized Clinical Trial. JAMA Neurol. [Europe PMC free article] [Abstract] [Google Scholar]
  • Poddar R, Deb I, Mukherjee S, Paul S, 2010. NR2B-NMDA receptor mediated modulation of the tyrosine phosphatase STEP regulates glutamate induced neuronal cell death. J Neurochem 115, 1350–1362. [Europe PMC free article] [Abstract] [Google Scholar]
  • Poddar R, Rajagopal S, Shuttleworth CW, Paul S, 2016. Zn2+-dependent Activation of the Trk Signaling Pathway Induces Phosphorylation of the Brain-enriched Tyrosine Phosphatase STEP: MOLECULAR BASIS FOR ZN2+-INDUCED ERK MAPK ACTIVATION. J Biol Chem 291, 813–825. [Europe PMC free article] [Abstract] [Google Scholar]
  • Poddar R, Rajagopal S, Winter L, Allan AM, Paul S, 2019. A peptide mimetic of tyrosine phosphatase STEP as a potential therapeutic agent for treatment of cerebral ischemic stroke. J Cereb Blood Flow Metab 39, 1069–1084. [Europe PMC free article] [Abstract] [Google Scholar]
  • Prabhakaran S, Ruff I, Bernstein RA, 2015. Acute stroke intervention: a systematic review. JAMA 313, 1451–1462. [Abstract] [Google Scholar]
  • Pruefer D, Scalia R, Lefer AM, 1999. Simvastatin inhibits leukocyte-endothelial cell interactions and protects against inflammatory processes in normocholesterolemic rats. Arterioscler Thromb Vasc Biol 19, 2894–2900. [Abstract] [Google Scholar]
  • Pulido R, Zuniga A, Ullrich A, 1998. PTP-SL and STEP protein tyrosine phosphatases regulate the activation of the extracellular signal-regulated kinases ERK1 and ERK2 by association through a kinase interaction motif. EMBO J 17, 7337–7350. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pulsinelli W, 1992. Pathophysiology of acute ischaemic stroke. Lancet 339, 533–536. [Abstract] [Google Scholar]
  • Rajagopal S, Deb I, Poddar R, Paul S, 2016. Aging is associated with dimerization and inactivation of the brain-enriched tyrosine phosphatase STEP. Neurobiol Aging 41, 25–38. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ramos-Cabrer P, Campos F, Sobrino T, Castillo J, 2011. Targeting the ischemic penumbra. Stroke 42, S7–11. [Abstract] [Google Scholar]
  • Reading JL, Vaes B, Hull C, Sabbah S, Hayday T, Wang NS, DiPiero A, Lehman NA, Taggart JM, Carty F, English K, Pinxteren J, Deans R, Ting AE, Tree TIM, 2015. Suppression of IL-7-dependent Effector T-cell Expansion by Multipotent Adult Progenitor Cells and PGE2. Mol Ther 23, 1783–1793. [Europe PMC free article] [Abstract] [Google Scholar]
  • Reck M, Blais N, Juhasz E, Gorbunova V, Jones CM, Urban L, Orlov S, Barlesi F, Kio E, Keilholz U, Qin Q, Qian J, Nickner C, Dziubinski J, Xiong H, Mittapalli RK, Dunbar M, Ansell P, He L, McKee M, Giranda V, Ramalingam SS, 2017. Smoking History Predicts Sensitivity to PARP Inhibitor Veliparib in Patients with Advanced Non-Small Cell Lung Cancer. J Thorac Oncol 12, 1098–1108. [Abstract] [Google Scholar]
  • Relton JK, Sloan KE, Frew EM, Whalley ET, Adams SP, Lobb RR, 2001. Inhibition of alpha4 integrin protects against transient focal cerebral ischemia in normotensive and hypertensive rats. Stroke 32, 199–205. [Abstract] [Google Scholar]
  • Ren C, Yan Z, Wei D, Gao X, Chen X, Zhao H, 2009. Limb remote ischemic postconditioning protects against focal ischemia in rats. Brain Res 1288, 88–94. [Europe PMC free article] [Abstract] [Google Scholar]
  • Rezaie AR, 2010. Regulation of the protein C anticoagulant and anti-inflammatory pathways. Curr Med Chem 17, 2059–2069. [Europe PMC free article] [Abstract] [Google Scholar]
  • Rice GP, Hartung HP, Calabresi PA, 2005. Anti-alpha4 integrin therapy for multiple sclerosis: mechanisms and rationale. Neurology 64, 1336–1342. [Abstract] [Google Scholar]
  • Rikitake Y, Kim HH, Huang Z, Seto M, Yano K, Asano T, Moskowitz MA, Liao JK, 2005. Inhibition of Rho kinase (ROCK) leads to increased cerebral blood flow and stroke protection. Stroke 36, 2251–2257. [Europe PMC free article] [Abstract] [Google Scholar]
  • Rischke R, Krieglstein J, 1991. Protective effect of vinpocetine against brain damage caused by ischemia. Jpn J Pharmacol 56, 349–356. [Abstract] [Google Scholar]
  • Rolain H, Miranpuri G, Ahmed A, 2019. Edaravone’s antioxidant capabilities and its therapeutic benefits for post-ischemic stroke: a mini review. On J Complement & Alt Med 2. [Google Scholar]
  • Romano M, Diomede L, Sironi M, Massimiliano L, Sottocorno M, Polentarutti N, Guglielmotti A, Albani D, Bruno A, Fruscella P, Salmona M, Vecchi A, Pinza M, Mantovani A, 2000a. Inhibition of monocyte chemotactic protein-1 synthesis by statins. Lab Invest 80, 1095–1100. [Abstract] [Google Scholar]
  • Romano M, Mezzetti A, Marulli C, Ciabattoni G, Febo F, Di Ienno S, Roccaforte S, Vigneri S, Nubile G, Milani M, Davi G, 2000b. Fluvastatin reduces soluble P-selectin and ICAM-1 levels in hypercholesterolemic patients: role of nitric oxide. J Investig Med 48, 183–189. [Abstract] [Google Scholar]
  • Romanos E, Planas AM, Amaro S, Chamorro A, 2007. Uric acid reduces brain damage and improves the benefits of rt-PA in a rat model of thromboembolic stroke. J Cereb Blood Flow Metab 27, 14–20. [Abstract] [Google Scholar]
  • Roy MW, Dempsey RJ, Meyer KL, Donaldson DL, Tibbs PA, Young AB, 1985. Effects of verapamil and diltiazem on acute stroke in cats. J Neurosurg 63, 929–936. [Abstract] [Google Scholar]
  • Rubbert-Roth A, Furst DE, Nebesky JM, Jin A, Berber E, 2018. A Review of Recent Advances Using Tocilizumab in the Treatment of Rheumatic Diseases. Rheumatol Ther 5, 21–42. [Europe PMC free article] [Abstract] [Google Scholar]
  • Rudick RA, Sandrock A, 2004. Natalizumab: alpha 4-integrin antagonist selective adhesion molecule inhibitors for MS. Expert Rev Neurother 4, 571–580. [Abstract] [Google Scholar]
  • Sacchetti ML, 2008. Is it time to definitely abandon neuroprotection in acute ischemic stroke? Stroke 39, 1659–1660. [Abstract] [Google Scholar]
  • Safarian F, Khallaghi B, Ahmadiani A, Dargahi L, 2015. Activation of S1P(1) receptor regulates PI3K/Akt/FoxO3a pathway in response to oxidative stress in PC12 cells. J Mol Neurosci 56, 177–187. [Abstract] [Google Scholar]
  • Santos MS, Duarte AI, Moreira PI, Oliveira CR, 2000. Synaptosomal response to oxidative stress: effect of vinpocetine. Free Radic Res 32, 57–66. [Abstract] [Google Scholar]
  • Sarfo FS, Ovbiagele B, Gebregziabher M, Wahab K, Akinyemi R, Akpalu A, Akpa O, Obiako R, Owolabi L, Jenkins C, Owolabi M, Siren, 2018. Stroke Among Young West Africans: Evidence From the SIREN (Stroke Investigative Research and Educational Network) Large Multisite Case-Control Study. Stroke 49, 1116–1122. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sato M, Tani E, Fujikawa H, Kaibuchi K, 2000. Involvement of Rho-kinase-mediated phosphorylation of myosin light chain in enhancement of cerebral vasospasm. Circ Res 87, 195–200. [Abstract] [Google Scholar]
  • Satoh S, Ikegaki I, Suzuki Y, Asano T, Shibuya M, Hidaka H, 1996. Neuroprotective properties of a protein kinase inhibitor against ischaemia-induced neuronal damage in rats and gerbils. Br J Pharmacol 118, 1592–1596. [Europe PMC free article] [Abstract] [Google Scholar]
  • Satoh S, Kobayashi T, Hitomi A, Ikegaki I, Suzuki Y, Shibuya M, Yoshida J, Asano T, 1999. Inhibition of neutrophil migration by a protein kinase inhibitor for the treatment of ischemic brain infarction. Jpn J Pharmacol 80, 41–48. [Abstract] [Google Scholar]
  • Satoh S, Yamaguchi T, Hitomi A, Sato N, Shiraiwa K, Ikegaki I, Asano T, Shimokawa H, 2002. Fasudil attenuates interstitial fibrosis in rat kidneys with unilateral ureteral obstruction. Eur J Pharmacol 455, 169–174. [Abstract] [Google Scholar]
  • Sattler R, Xiong Z, Lu WY, Hafner M, MacDonald JF, Tymianski M, 1999. Specific coupling of NMDA receptor activation to nitric oxide neurotoxicity by PSD-95 protein. Science 284, 1845–1848. [Abstract] [Google Scholar]
  • Sauer D, Rischke R, Beck T, Rossberg C, Mennel HD, Bielenberg GW, Krieglstein J, 1988. Vinpocetine prevents ischemic cell damage in rat hippocampus. Life Sci 43, 1733–1739. [Abstract] [Google Scholar]
  • Saver JL, 2013. The 2012 Feinberg Lecture: treatment swift and treatment sure. Stroke 44, 270–277. [Europe PMC free article] [Abstract] [Google Scholar]
  • Savica V, Calo LA, Santoro D, Monardo P, Mallamace A, Bellinghieri G, 2011. Urine therapy through the centuries. J Nephrol 24 Suppl 17, S123–125. [Abstract] [Google Scholar]
  • Schachter M, 2005. Chemical, pharmacokinetic and pharmacodynamic properties of statins: an update. Fundam Clin Pharmacol 19, 117–125. [Abstract] [Google Scholar]
  • Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S, 2011. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta 1813, 878–888. [Abstract] [Google Scholar]
  • Sheth KN, Elm JJ, Molyneaux BJ, Hinson H, Beslow LA, Sze GK, Ostwaldt AC, Del Zoppo GJ, Simard JM, Jacobson S, Kimberly WT, 2016. Safety and efficacy of intravenous glyburide on brain swelling after large hemispheric infarction (GAMES-RP): a randomised, doubleblind, placebo-controlled phase 2 trial. Lancet Neurol 15, 1160–1169. [Abstract] [Google Scholar]
  • Sheth KN, Petersen NH, Cheung K, Elm JJ, Hinson HE, Molyneaux BJ, Beslow LA, Sze GK, Simard JM, Kimberly WT, 2018. Long-Term Outcomes in Patients Aged </=70 Years With Intravenous Glyburide From the Phase II GAMES-RP Study of Large Hemispheric Infarction: An Exploratory Analysis. Stroke 49, 1457–1463. [Europe PMC free article] [Abstract] [Google Scholar]
  • Shibata H, Arai S, Izawa M, Murasaki M, Takamatsu Y, Izawa O, Takahashi C, Tanaka M, 1998. Phase I clinical study of MCI-186 (Edaravone, 3-methyl-1-phenyl-2-pyrazolin-5-one) in healthy volunteers: safety and pharmacokinetics of single and multiple administrations. Jpn J Clin Pharmacol Ther 29, 863–876. [Google Scholar]
  • Shibata M, Kumar SR, Amar A, Fernandez JA, Hofman F, Griffin JH, Zlokovic BV, 2001. Anti-inflammatory, antithrombotic, and neuroprotective effects of activated protein C in a murine model of focal ischemic stroke. Circulation 103, 1799–1805. [Abstract] [Google Scholar]
  • Shibuya M, Hirai S, Seto M, Satoh S, Ohtomo E, Fasudil Ischemic Stroke Study, G., 2005. Effects of fasudil in acute ischemic stroke: results of a prospective placebo-controlled double-blind trial. J Neurol Sci 238, 31–39. [Abstract] [Google Scholar]
  • Shichinohe H, Kuroda S, Yasuda H, Ishikawa T, Iwai M, Horiuchi M, Iwasaki Y, 2004. Neuroprotective effects of the free radical scavenger Edaravone (MCI-186) in mice permanent focal brain ischemia. Brain Res 1029, 200–206. [Abstract] [Google Scholar]
  • Shimokawa H, Seto M, Katsumata N, Amano M, Kozai T, Yamawaki T, Kuwata K, Kandabashi T, Egashira K, Ikegaki I, Asano T, Kaibuchi K, Takeshita A, 1999. Rho-kinasemediated pathway induces enhanced myosin light chain phosphorylations in a swine model of coronary artery spasm. Cardiovasc Res 43, 1029–1039. [Abstract] [Google Scholar]
  • Shinohara Y, Saito I, Kobayashi S, Uchiyama S, 2009. Edaravone (radical scavenger) versus sodium ozagrel (antiplatelet agent) in acute noncardioembolic ischemic stroke (EDO trial). Cerebrovasc Dis 27, 485–492. [Abstract] [Google Scholar]
  • Shu ZM, Shu XD, Li HQ, Sun Y, Shan H, Sun XY, Du RH, Lu M, Xiao M, Ding JH, Hu G, 2016. Ginkgolide B Protects Against Ischemic Stroke Via Modulating Microglia Polarization in Mice. CNS Neurosci Ther 22, 729–739. [Europe PMC free article] [Abstract] [Google Scholar]
  • Simard JM, Chen M, Tarasov KV, Bhatta S, Ivanova S, Melnitchenko L, Tsymbalyuk N, West GA, Gerzanich V, 2006. Newly expressed SUR1-regulated NC(Ca-ATP) channel mediates cerebral edema after ischemic stroke. Nat Med 12, 433–440. [Europe PMC free article] [Abstract] [Google Scholar]
  • Simard JM, Tsymbalyuk N, Tsymbalyuk O, Ivanova S, Yurovsky V, Gerzanich V, 2010. Glibenclamide is superior to decompressive craniectomy in a rat model of malignant stroke. Stroke 41, 531–537. [Europe PMC free article] [Abstract] [Google Scholar]
  • Simard JM, Woo SK, Tsymbalyuk N, Voloshyn O, Yurovsky V, Ivanova S, Lee R, Gerzanich V, 2012. Glibenclamide-10-h Treatment Window in a Clinically Relevant Model of Stroke. Transl Stroke Res 3, 286–295. [Europe PMC free article] [Abstract] [Google Scholar]
  • Simard JM, Yurovsky V, Tsymbalyuk N, Melnichenko L, Ivanova S, Gerzanich V, 2009. Protective effect of delayed treatment with low-dose glibenclamide in three models of ischemic stroke. Stroke 40, 604–609. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sironi L, Banfi C, Brioschi M, Gelosa P, Guerrini U, Nobili E, Gianella A, Paoletti R, Tremoli E, Cimino M, 2006. Activation of NF-kB and ERK1/2 after permanent focal ischemia is abolished by simvastatin treatment. Neurobiol Dis 22, 445–451. [Abstract] [Google Scholar]
  • Sironi L, Cimino M, Guerrini U, Calvio AM, Lodetti B, Asdente M, Balduini W, Paoletti R, Tremoli E, 2003. Treatment with statins after induction of focal ischemia in rats reduces the extent of brain damage. Arterioscler Thromb Vasc Biol 23, 322–327. [Abstract] [Google Scholar]
  • Sitges M, Galvan E, Nekrassov V, 2005. Vinpocetine blockade of sodium channels inhibits the rise in sodium and calcium induced by 4-aminopyridine in synaptosomes. Neurochem Int 46, 533–540. [Abstract] [Google Scholar]
  • Sitges M, Sanchez-Tafolla BM, Chiu LM, Aldana BI, Guarneros A, 2011. Vinpocetine inhibits glutamate release induced by the convulsive agent 4-aminopyridine more potently than several antiepileptic drugs. Epilepsy Res 96, 257–266. [Abstract] [Google Scholar]
  • Smith CJ, Emsley HC, Gavin CM, Georgiou RF, Vail A, Barberan EM, del Zoppo GJ, Hallenbeck JM, Rothwell NJ, Hopkins SJ, Tyrrell PJ, 2004. Peak plasma interleukin-6 and other peripheral markers of inflammation in the first week of ischaemic stroke correlate with brain infarct volume, stroke severity and long-term outcome. BMC Neurol 4, 2. [Europe PMC free article] [Abstract] [Google Scholar]
  • Smith WS, Sung G, Saver J, Budzik R, Duckwiler G, Liebeskind DS, Lutsep HL, Rymer MM, Higashida RT, Starkman S, Gobin YP, Multi MI, Frei D, Grobelny T, Hellinger F, Huddle D, Kidwell C, Koroshetz W, Marks M, Nesbit G, Silverman IE, 2008. Mechanical thrombectomy for acute ischemic stroke: final results of the Multi MERCI trial. Stroke 39, 1205–1212. [Abstract] [Google Scholar]
  • Snyder EM, Nong Y, Almeida CG, Paul S, Moran T, Choi EY, Nairn AC, Salter MW, Lombroso PJ, Gouras GK, Greengard P, 2005. Regulation of NMDA receptor trafficking by amyloid-beta. Nat Neurosci 8, 1051–1058. [Abstract] [Google Scholar]
  • Soriano FX, Martel MA, Papadia S, Vaslin A, Baxter P, Rickman C, Forder J, Tymianski M, Duncan R, Aarts M, Clarke P, Wyllie DJ, Hardingham GE, 2008. Specific targeting of pro-death NMDA receptor signals with differing reliance on the NR2B PDZ ligand. J Neurosci 28, 10696–10710. [Europe PMC free article] [Abstract] [Google Scholar]
  • Squadrito GL, Cueto R, Splenser AE, Valavanidis A, Zhang H, Uppu RM, Pryor WA, 2000. Reaction of uric acid with peroxynitrite and implications for the mechanism of neuroprotection by uric acid. Arch Biochem Biophys 376, 333–337. [Abstract] [Google Scholar]
  • Stromgaard K, Nakanishi K, 2004. Chemistry and biology of terpene trilactones from Ginkgo biloba. Angew Chem Int Ed Engl 43, 1640–1658. [Abstract] [Google Scholar]
  • Strosznajder RP, Czubowicz K, Jesko H, Strosznajder JB, 2010. Poly(ADP-ribose) metabolism in brain and its role in ischemia pathology. Mol Neurobiol 41, 187–196. [Abstract] [Google Scholar]
  • Stuart RM, Helbok R, Kurtz P, Schmidt M, Fernandez L, Lee K, Badjatia N, Mayer SA, Lavine S, Meyers P, Connolly ES, Claassen J, 2011. High-dose intra-arterial verapamil for the treatment of cerebral vasospasm after subarachnoid hemorrhage: prolonged effects on hemodynamic parameters and brain metabolism. Neurosurgery 68, 337–345; discussion 345. [Abstract] [Google Scholar]
  • Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, Gao Y, Pietras K, Mann K, Yepes M, Strickland DK, Betsholtz C, Eriksson U, Lawrence DA, 2008. Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat Med 14, 731–737. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sun D, Lee YS, Malhotra A, Kim HK, Matecic M, Evans C, Jensen RV, Moskaluk CA, Dutta A, 2011a. miR-99 family of MicroRNAs suppresses the expression of prostate-specific antigen and prostate cancer cell proliferation. Cancer Res 71, 1313–1324. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sun HS, Doucette TA, Liu Y, Fang Y, Teves L, Aarts M, Ryan CL, Bernard PB, Lau A, Forder JP, Salter MW, Wang YT, Tasker RA, Tymianski M, 2008. Effectiveness of PSD95 inhibitors in permanent and transient focal ischemia in the rat. Stroke 39, 2544–2553. [Abstract] [Google Scholar]
  • Sun J, Li Y, Fang W, Mao L, 2011b. Therapeutic time window for treatment of focal cerebral ischemia reperfusion injury with XQ-1h in rats. Eur J Pharmacol 666, 105–110. [Abstract] [Google Scholar]
  • Sun J, Tong L, Luan Q, Deng J, Li Y, Li Z, Dong H, Xiong L, 2012. Protective effect of delayed remote limb ischemic postconditioning: role of mitochondrial K(ATP) channels in a rat model of focal cerebral ischemic reperfusion injury. J Cereb Blood Flow Metab 32, 851–859. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sun MS, Jin H, Sun X, Huang S, Zhang FL, Guo ZN, Yang Y, 2018. Free Radical Damage in Ischemia-Reperfusion Injury: An Obstacle in Acute Ischemic Stroke after Revascularization Therapy. Oxid Med Cell Longev 2018, 3804979. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sun Y, Cheng X, Wang H, Mu X, Liang Y, Luo Y, Qu H, Zhao C, 2017. dl-3-n-butylphthalide promotes neuroplasticity and motor recovery in stroke rats. Behav Brain Res 329, 67–74. [Abstract] [Google Scholar]
  • Sun Y, Gui H, Li Q, Luo ZM, Zheng MJ, Duan JL, Liu X, 2013. MicroRNA-124 protects neurons against apoptosis in cerebral ischemic stroke. CNS Neurosci Ther 19, 813–819. [Europe PMC free article] [Abstract] [Google Scholar]
  • Swindell WR, Bojanowski K, Kindy MS, Chau RMW, Ko D, 2018. GM604 regulates developmental neurogenesis pathways and the expression of genes associated with amyotrophic lateral sclerosis. Transl Neurodegener 7, 30. [Europe PMC free article] [Abstract] [Google Scholar]
  • Takahashi K, Pieper AA, Croul SE, Zhang J, Snyder SH, Greenberg JH, 1999. Posttreatment with an inhibitor of poly(ADP-ribose) polymerase attenuates cerebral damage in focal ischemia. Brain Res 829, 46–54. [Abstract] [Google Scholar]
  • Tamagnan G, Tavares A, Barret O, Alagille D, Seibyl J, Marek K, Maguire R, Briard E, Schmouder R, Behrje R, Jennings D, 2012. Brain distribution of BZM055, an analog of fingolimod (FTY720), in human. Mult Scler 18, 379. [Google Scholar]
  • Tan KS, Armugam A, Sepramaniam S, Lim KY, Setyowati KD, Wang CW, Jeyaseelan K, 2009. Expression profile of MicroRNAs in young stroke patients. PLoS One 4, e7689. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tanaka S, Takehashi M, Iida S, Kitajima T, Kamanaka Y, Stedeford T, Banasik M, Ueda K, 2005. Mitochondrial impairment induced by poly(ADP-ribose) polymerase-1 activation in cortical neurons after oxygen and glucose deprivation. J Neurochem 95, 179–190. [Abstract] [Google Scholar]
  • Tanaka T, Narazaki M, Ogata A, Kishimoto T, 2014. A new era for the treatment of inflammatory autoimmune diseases by interleukin-6 blockade strategy. Semin Immunol 26, 88–96. [Abstract] [Google Scholar]
  • Tao Z, Zhao H, Wang R, Liu P, Yan F, Zhang C, Ji X, Luo Y, 2015. Neuroprotective effect of microRNA-99a against focal cerebral ischemia-reperfusion injury in mice. J Neurol Sci 355, 113–119. [Abstract] [Google Scholar]
  • Teramoto S, Miyamoto N, Yatomi K, Tanaka Y, Oishi H, Arai H, Hattori N, Urabe T, 2011. Exendin-4, a glucagon-like peptide-1 receptor agonist, provides neuroprotection in mice transient focal cerebral ischemia. J Cereb Blood Flow Metab 31, 1696–1705. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tubridy N, Behan PO, Capildeo R, Chaudhuri A, Forbes R, Hawkins CP, Hughes RA, Palace J, Sharrack B, Swingler R, Young C, Moseley IF, MacManus DG, Donoghue S, Miller DH, 1999. The effect of anti-alpha4 integrin antibody on brain lesion activity in MS. The UK Antegren Study Group. Neurology 53, 466–472. [Abstract] [Google Scholar]
  • Tuo Y, Liu Z, Feng L, Shi Z, 2017. MicroRNA-29a protects against cerebral ischemia and reperfusion injury by targeting NADPH Oxidase 4. Stroke 48, AWMP37. [Google Scholar]
  • Ueda T, Yamamoto YL, Diksic M, 1989. Transvenous perfusion of the brain with verapamil during focal cerebral ischemia in rats. Stroke 20, 501–506. [Abstract] [Google Scholar]
  • Valko K, Kindy M, Evans J, Ko D, 2018. In vitro biomimetic HPLC and in vivo characterisation of GM6, an endogenous regulator peptide drug candidate for amyotrophic lateral sclerosis. ADMET & DMPK 6, 176–189. [Google Scholar]
  • van Beek TA, 2005. Ginkgolides and bilobalide: their physical, chromatographic and spectroscopic properties. Bioorg Med Chem 13, 5001–5012. [Abstract] [Google Scholar]
  • Vaughan CJ, Gotto AM Jr., Basson CT, 2000. The evolving role of statins in the management of atherosclerosis. J Am Coll Cardiol 35, 1–10. [Abstract] [Google Scholar]
  • Veltkamp R, Gill D, 2016. Clinical Trials of Immunomodulation in Ischemic Stroke. Neurotherapeutics 13, 791–800. [Europe PMC free article] [Abstract] [Google Scholar]
  • Virley D, Beech JS, Smart SC, Williams SC, Hodges H, Hunter AJ, 2000. A temporal MRI assessment of neuropathology after transient middle cerebral artery occlusion in the rat: correlations with behavior. J Cereb Blood Flow Metab 20, 563–582. [Abstract] [Google Scholar]
  • Wahlgren N, Thoren M, Hojeberg B, Kall TB, Laska AC, Sjostrand C, Hoijer J, Almqvist H, Holmin S, Lilja A, Fredriksson L, Lawrence D, Eriksson U, Ahmed N, 2017. Randomized assessment of imatinib in patients with acute ischaemic stroke treated with intravenous thrombolysis. J Intern Med 281, 273–283. [Europe PMC free article] [Abstract] [Google Scholar]
  • Waje-Andreassen U, Krakenes J, Ulvestad E, Thomassen L, Myhr KM, Aarseth J, Vedeler CA, 2005. IL-6: an early marker for outcome in acute ischemic stroke. Acta Neurol Scand 111, 360–365. [Abstract] [Google Scholar]
  • Walker PA, Bedi SS, Shah SK, Jimenez F, Xue H, Hamilton JA, Smith P, Thomas CP, Mays RW, Pati S, Cox CS Jr., 2012. Intravenous multipotent adult progenitor cell therapy after traumatic brain injury: modulation of the resident microglia population. J Neuroinflammation 9, 228. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang A, Chau R, Chow S, Zhang Z, Li Z, 1995. Effects of myogenic 22 and 35kD neurotrophic factors on axonal regeneration in free peripheral autografts into rat spinal cord. Chinese Journal of Spine and Spinal Cord 5, 248–252. [Google Scholar]
  • Wang H, Zhang K, Zhao L, Tang J, Gao L, Wei Z, 2014. Anti-inflammatory effects of vinpocetine on the functional expression of nuclear factor-kappa B and tumor necrosis factor-alpha in a rat model of cerebral ischemia-reperfusion injury. Neurosci Lett 566, 247–251. [Abstract] [Google Scholar]
  • Wang Q, Tang XN, Yenari MA, 2007. The inflammatory response in stroke. J Neuroimmunol 184, 53–68. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang S, Ma F, Huang L, Zhang Y, Peng Y, Xing C, Feng Y, Wang X, Peng Y, 2018. Dl-3n-Butylphthalide (NBP): A Promising Therapeutic Agent for Ischemic Stroke. CNS Neurol Disord Drug Targets 17, 338–347. [Abstract] [Google Scholar]
  • Wang SW, Sun YM, 2014. The IL-6/JAK/STAT3 pathway: potential therapeutic strategies in treating colorectal cancer (Review). Int J Oncol 44, 1032–1040. [Abstract] [Google Scholar]
  • Wang Y, Thiyagarajan M, Chow N, Singh I, Guo H, Davis TP, Zlokovic BV, 2009. Differential neuroprotection and risk for bleeding from activated protein C with varying degrees of anticoagulant activity. Stroke 40, 1864–1869. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang Y, Zhang Z, Chow N, Davis TP, Griffin JH, Chopp M, Zlokovic BV, 2012. An activated protein C analog with reduced anticoagulant activity extends the therapeutic window of tissue plasminogen activator for ischemic stroke in rodents. Stroke 43, 2444–2449. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang Y, Zhao Z, Chow N, Rajput PS, Griffin JH, Lyden PD, Zlokovic BV, 2013. Activated protein C analog protects from ischemic stroke and extends the therapeutic window of tissue-type plasminogen activator in aged female mice and hypertensive rats. Stroke 44, 3529–3536. [Europe PMC free article] [Abstract] [Google Scholar]
  • Watanabe T, Yuki S, Egawa M, Nishi H, 1994. Protective effects of MCI-186 on cerebral ischemia: possible involvement of free radical scavenging and antioxidant actions. J Pharmacol Exp Ther 268, 1597–1604. [Abstract] [Google Scholar]
  • Wauquier A, Melis W, Janssen PA, 1989. Long-term neurological assessment of the postresuscitative effects of flunarizine, verapamil and nimodipine in a new model of global complete ischaemia. Neuropharmacology 28, 837–846. [Abstract] [Google Scholar]
  • Wei D, Ren C, Chen X, Zhao H, 2012. The chronic protective effects of limb remote preconditioning and the underlying mechanisms involved in inflammatory factors in rat stroke. PLoS One 7, e30892. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wei Y, Yemisci M, Kim HH, Yung LM, Shin HK, Hwang SK, Guo S, Qin T, Alsharif N, Brinkmann V, Liao JK, Lo EH, Waeber C, 2011. Fingolimod provides long-term protection in rodent models of cerebral ischemia. Ann Neurol 69, 119–129. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wei Z, Lyu Y, Yang X, Chen X, Zhong P, Wu D, 2018. Therapeutic Values of Human Urinary Kallidinogenase on Cerebrovascular Diseases. Front Neurol 9, 403. [Europe PMC free article] [Abstract] [Google Scholar]
  • Weitz-Schmidt G, 2002. Statins as anti-inflammatory agents. Trends Pharmacol Sci 23, 482–486. [Abstract] [Google Scholar]
  • Williams PD, Zlokovic BV, Griffin JH, Pryor KE, Davis TP, 2012. Preclinical safety and pharmacokinetic profile of 3K3A-APC, a novel, modified activated protein C for ischemic stroke. Curr Pharm Des 18, 4215–4222. [Europe PMC free article] [Abstract] [Google Scholar]
  • Willis MA, Cohen JA, 2013. Fingolimod therapy for multiple sclerosis. Semin Neurol 33, 37–44. [Abstract] [Google Scholar]
  • Won S, Incontro S, Li Y, Nicoll RA, Roche KW, 2019. The STEP61 interactome reveals subunit-specific AMPA receptor binding and synaptic regulation. Proc Natl Acad Sci U S A 116, 8028–8037. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wong CM, Wang Y, Lee JT, Huang Z, Wu D, Xu A, Lam KS, 2014. Adropin is a brain membrane-bound protein regulating physical activity via the NB-3/Notch signaling pathway in mice. J Biol Chem 289, 25976–25986. [Europe PMC free article] [Abstract] [Google Scholar]
  • Woo SK, Kwon MS, Ivanov A, Gerzanich V, Simard JM, 2013. The sulfonylurea receptor 1 (Sur1)-transient receptor potential melastatin 4 (Trpm4) channel. J Biol Chem 288, 3655–3667. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wu D, Lyu Y, Zhong P, Liu F, Liu X, 2017a. Human Urinary kallidinogenase promotes good recovery in ischemic stroke patients with level 3 hypertension. Brain Behav 7, e00752. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wu LR, Liu L, Xiong XY, Zhang Q, Wang FX, Gong CX, Zhong Q, Yang YR, Meng ZY, Yang QW, 2017b. Vinpocetine alleviate cerebral ischemia/reperfusion injury by downregulating TLR4/MyD88/NF-kappaB signaling. Oncotarget 8, 80315–80324. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wu TW, Zeng LH, Wu J, Fung KP, 2000. MCI-186: further histochemical and biochemical evidence of neuroprotection. Life Sci 67, 2387–2392. [Abstract] [Google Scholar]
  • Xiang W, He J, Huang C, Chen L, Tao D, Wu X, Wang M, Luo G, Xiao X, Zeng F, Jiang G, 2015. miR-106b-5p targets tumor suppressor gene SETD2 to inactive its function in clear cell renal cell carcinoma. Oncotarget 6, 4066–4079. [Europe PMC free article] [Abstract] [Google Scholar]
  • Xiong XY, Liu L, Yang QW, 2018. Refocusing Neuroprotection in Cerebral Reperfusion Era: New Challenges and Strategies. Front Neurol 9, 249. [Europe PMC free article] [Abstract] [Google Scholar]
  • Xu J, Kurup P, Bartos JA, Patriarchi T, Hell JW, Lombroso PJ, 2012. Striatal-enriched protein-tyrosine phosphatase (STEP) regulates Pyk2 kinase activity. J Biol Chem 287, 20942–20956. [Europe PMC free article] [Abstract] [Google Scholar]
  • Xu LJ, Ouyang YB, Xiong X, Stary CM, Giffard RG, 2015. Post-stroke treatment with miR181 antagomir reduces injury and improves long-term behavioral recovery in mice after focal cerebral ischemia. Exp Neurol 264, 1–7. [Europe PMC free article] [Abstract] [Google Scholar]
  • Xu ZQ, Zhou Y, Shao BZ, Zhang JJ, Liu C, 2019. A Systematic Review of Neuroprotective Efficacy and Safety of DL-3-N-Butylphthalide in Ischemic Stroke. Am J Chin Med 47, 507–525. [Abstract] [Google Scholar]
  • Ya BL, Liu Q, Li HF, Cheng HJ, Yu T, Chen L, Wang Y, Yuan LL, Li WJ, Liu WY, Bai B, 2018. Uric Acid Protects against Focal Cerebral Ischemia/Reperfusion-Induced Oxidative Stress via Activating Nrf2 and Regulating Neurotrophic Factor Expression. Oxid Med Cell Longev 2018, 6069150. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yagi K, Kitazato KT, Uno M, Tada Y, Kinouchi T, Shimada K, Nagahiro S, 2009. Edaravone, a free radical scavenger, inhibits MMP-9-related brain hemorrhage in rats treated with tissue plasminogen activator. Stroke 40, 626–631. [Abstract] [Google Scholar]
  • Yamamoto T, Yuki S, Watanabe T, Mitsuka M, Saito KI, Kogure K, 1997. Delayed neuronal death prevented by inhibition of increased hydroxyl radical formation in a transient cerebral ischemia. Brain Res 762, 240–242. [Abstract] [Google Scholar]
  • Yamashita T, Sawamoto K, Suzuki S, Suzuki N, Adachi K, Kawase T, Mihara M, Ohsugi Y, Abe K, Okano H, 2005. Blockade of interleukin-6 signaling aggravates ischemic cerebral damage in mice: possible involvement of Stat3 activation in the protection of neurons. J Neurochem 94, 459–468. [Abstract] [Google Scholar]
  • Yan RY, Wang SJ, Yao GT, Liu ZG, Xiao N, 2017. The protective effect and its mechanism of 3-n-butylphthalide pretreatment on cerebral ischemia reperfusion injury in rats. Eur Rev Med Pharmacol Sci 21, 5275–5282. [Abstract] [Google Scholar]
  • Yang B, Hamilton JA, Valenzuela KS, Bogaerts A, Xi X, Aronowski J, Mays RW, Savitz SI, 2017a. Multipotent Adult Progenitor Cells Enhance Recovery After Stroke by Modulating the Immune Response from the Spleen. Stem Cells 35, 1290–1302. [Abstract] [Google Scholar]
  • Yang C, DeMars K, Candelario-Jalil E, 2017b. Adropin is profoundly neuroprotective in experimental ischemic stroke. Stroke 48, ATP277. [Google Scholar]
  • Yang C, DeMars KM, Hawkins KE, Candelario-Jalil E, 2016. Adropin reduces paracellular permeability of rat brain endothelial cells exposed to ischemia-like conditions. Peptides 81, 29–37. [Abstract] [Google Scholar]
  • Yang C, Sanz Y, DeMars K, Butler A, Candelario-Jalil E, 2020. Neuroprotective effects of endogenous adropin in experimental ischemic stroke. Stroke 51, AWP315. [Google Scholar]
  • Yang CS, Guo A, Li Y, Shi K, Shi FD, Li M, 2019a. Dl-3-n-butylphthalide Reduces Neurovascular Inflammation and Ischemic Brain Injury in Mice. Aging Dis 10, 964–976. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yang J, Balkaya M, Beltran C, Heo JH, Cho S, 2019b. Remote Postischemic Conditioning Promotes Stroke Recovery by Shifting Circulating Monocytes to CCR2(+) Proinflammatory Subset. J Neurosci 39, 7778–7789. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yang J, Su J, Wan F, Yang N, Jiang H, Fang M, Xiao H, Wang J, Tang J, 2017c. Tissue kallikrein protects against ischemic stroke by suppressing TLR4/NF-kappaB and activating Nrf2 signaling pathway in rats. Exp Ther Med 14, 1163–1170. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yang X, Tang X, Sun P, Shi Y, Liu K, Hassan SH, Stetler RA, Chen J, Yin KJ, 2017d. MicroRNA-15a/16–1 Antagomir Ameliorates Ischemic Brain Injury in Experimental Stroke. Stroke 48, 1941–1947. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yasuhara T, Matsukawa N, Yu G, Xu L, Mays RW, Kovach J, Deans R, Hess DC, Carroll JE, Borlongan CV, 2006. Transplantation of cryopreserved human bone marrow-derived multipotent adult progenitor cells for neonatal hypoxic-ischemic injury: targeting the hippocampus. Rev Neurosci 17, 215–225. [Abstract] [Google Scholar]
  • Yoshida H, Sasaki K, Namiki Y, Sato N, Tada N, 2005. Edaravone, a novel radical scavenger, inhibits oxidative modification of low-density lipoprotein (LDL) and reverses oxidized LDLmediated reduction in the expression of endothelial nitric oxide synthase. Atherosclerosis 179, 97–102. [Abstract] [Google Scholar]
  • Yoshida H, Yanai H, Namiki Y, Fukatsu-Sasaki K, Furutani N, Tada N, 2006. Neuroprotective effects of edaravone: a novel free radical scavenger in cerebrovascular injury. CNS Drug Rev 12, 920. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yoshimura S, Uchida K, Daimon T, Takashima R, Kimura K, Morimoto T, Investigator AT, 2017. Randomized Controlled Trial of Early Versus Delayed Statin Therapy in Patients With Acute Ischemic Stroke: ASSORT Trial (Administration of Statin on Acute Ischemic Stroke Patient). Stroke 48, 3057–3063. [Abstract] [Google Scholar]
  • Yousef GM, Diamandis EP, 2001. The new human tissue kallikrein gene family: structure, function, and association to disease. Endocr Rev 22, 184–204. [Abstract] [Google Scholar]
  • Yu J, Zhu H, Ko D, Kindy MS, 2008. Motoneuronotrophic factor analog GM6 reduces infarct volume and behavioral deficits following transient ischemia in the mouse. Brain Res 1238, 143–153. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yu ZF, Bruce-Keller AJ, Goodman Y, Mattson MP, 1998. Uric acid protects neurons against excitotoxic and metabolic insults in cell culture, and against focal ischemic brain injury in vivo. J Neurosci Res 53, 613–625. [Abstract] [Google Scholar]
  • Yuan M, Siegel C, Zeng Z, Li J, Liu F, McCullough LD, 2009. Sex differences in the response to activation of the poly (ADP-ribose) polymerase pathway after experimental stroke. Exp Neurol 217, 210–218. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zeng Z, Zhu J, Chen L, Wen W, Yu R, 2013. Biosynthesis pathways of ginkgolides. Pharmacogn Rev 7, 47–52. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhang C, Tao W, Liu M, Wang D, 2012a. Efficacy and safety of human urinary kallidinogenase injection for acute ischemic stroke: a systematic review. J Evid Based Med 5, 31–39. [Abstract] [Google Scholar]
  • Zhang C, Zang Y, Song Q, Zhao W, Li H, Hu L, Zhang Q, Gu F, Zhang C, 2020. Effects of butylphthalide injection on treatment of transient ischemic attack as shown by diffusion-weighted magnetic resonance imaging abnormality. Int J Neurosci 130, 454–460. [Abstract] [Google Scholar]
  • Zhang C, Zhao S, Zang Y, Gu F, Mao S, Feng S, Hu L, Zhang C, 2017. The efficacy and safety of Dl-3n-butylphthalide on progressive cerebral infarction: A randomized controlled STROBE study. Medicine (Baltimore) 96, e7257. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhang L, Yu WH, Wang YX, Wang C, Zhao F, Qi W, Chan WM, Huang Y, Wai MS, Dong J, Yew DT, 2012b. DL-3-n-Butylphthalide, an anti-oxidant agent, prevents neurological deficits and cerebral injury following stroke per functional analysis, magnetic resonance imaging and histological assessment. Curr Neurovasc Res 9, 167–175. [Abstract] [Google Scholar]
  • Zhang N, Komine-Kobayashi M, Tanaka R, Liu M, Mizuno Y, Urabe T, 2005. Edaravone reduces early accumulation of oxidative products and sequential inflammatory responses after transient focal ischemia in mice brain. Stroke 36, 2220–2225. [Abstract] [Google Scholar]
  • Zhang YS, Li JD, Yan C, 2018. An update on vinpocetine: New discoveries and clinical implications. Eur J Pharmacol 819, 30–34. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhao H, Tao Z, Wang R, Liu P, Yan F, Li J, Zhang C, Ji X, Luo Y, 2014. MicroRNA-23a3p attenuates oxidative stress injury in a mouse model of focal cerebral ischemia-reperfusion. Brain Res 1592, 65–72. [Abstract] [Google Scholar]
  • Zhao H, Wang J, Gao L, Wang R, Liu X, Gao Z, Tao Z, Xu C, Song J, Ji X, Luo Y, 2013. MiRNA-424 protects against permanent focal cerebral ischemia injury in mice involving suppressing microglia activation. Stroke 44, 1706–1713. [Abstract] [Google Scholar]
  • Zhao M, Hou S, Feng L, Shen P, Nan D, Zhang Y, Wang F, Ma D, Feng J, 2020. Vinpocetine Protects Against Cerebral Ischemia-Reperfusion Injury by Targeting Astrocytic Connexin43 via the PI3K/AKT Signaling Pathway. Front Neurosci 14, 223. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhao W, Che R, Li S, Ren C, Li C, Wu C, Lu H, Chen J, Duan J, Meng R, Ji X, 2018. Remote ischemic conditioning for acute stroke patients treated with thrombectomy. Ann Clin Transl Neurol 5, 850–856. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhou G, Li MH, Tudor G, Lu HT, Kadirvel R, Kallmes D, 2018. Remote Ischemic Conditioning in Cerebral Diseases and Neurointerventional Procedures: Recent Research Progress. Front Neurol 9, 339. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhou M, Huang Z, Wu X, Lu N, Rao X, 1994. Immunohistochemical localization of Motoneuronotrophic factor in fetal and neonatal rats. Acta Anatomica Sinica 25, 189–192. [Google Scholar]
  • Zhou M, Wu X, Chen S, 1997. Distribution of MNTF1 in spinal cord and limb muscles of mice with motoneuron disease. Acta Academiae Medicinae Sinicae 19, 171–178. [Abstract] [Google Scholar]
  • Zhou M, Yu W, Reb F, 1993. Changes in MNTF and its receptor in tongue muscle postdenervation of the hypoglossal nerve. Acta Anatomica Sinica 24, 391–395. [Google Scholar]
  • Zhou Q, Liao JK, 2009. Statins and cardiovascular diseases: from cholesterol lowering to pleiotropy. Curr Pharm Des 15, 467–478. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhou X, Wang HY, Wu B, Cheng CY, Xiao W, Wang ZZ, Yang YY, Li P, Yang H, 2017. Ginkgolide K attenuates neuronal injury after ischemic stroke by inhibiting mitochondrial fission and GSK-3beta-dependent increases in mitochondrial membrane permeability. Oncotarget 8, 44682–44693. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhu Z, Fu Y, Tian D, Sun N, Han W, Chang G, Dong Y, Xu X, Liu Q, Huang D, Shi FD, 2015. Combination of the Immune Modulator Fingolimod With Alteplase in Acute Ischemic Stroke: A Pilot Trial. Circulation 132, 1104–1112. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zlokovic BV, Griffin JH, 2011. Cytoprotective protein C pathways and implications for stroke and neurological disorders. Trends Neurosci 34, 198–209. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zlokovic BV, Zhang C, Liu D, Fernandez J, Griffin JH, Chopp M, 2005. Functional recovery after embolic stroke in rodents by activated protein C. Ann Neurol 58, 474–477. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/111835317
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/111835317

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1016/j.expneurol.2020.113518

Supporting
Mentioning
Contrasting
0
140
0

Article citations


Go to all (181) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

NINDS NIH HHS (3)

National Institutes of Health