Skip to main content
. 2016 Nov 25;17(12):1967. doi: 10.3390/ijms17121967

Table 5.

Anti-inflammatory treatments in ischemic stroke: evidence from preclinical studies.

Study Year Treatment Sample Size Outcome
IL-1Ra
Garcia et al. [256] 1995 13 with pMCAO and treated with IL-1Ra; 13 with pMCAO and treated with CSE buffer or placebo group (n = 13); 2 sham-operated animals treated with IL-1Ra or CSE (n = 2) 30 outbred male Wistar rats and fed Agway rat chow during the 4–6 quarantine days IL-1Ra in rats with pMCAO significantly decreased the number of necrotic neurons both at 24 h and 7 days after the arterial occlusion (p < 0.0001). Neurological scores were also significantly improved with and a non-significant decrease in the number of PMN leukocytes in the ischemic hemisphere was observed.
Yamasaki et al. [247] 1995 60 min of tMCAO followed by reperfusion; first IL-1β, then anti-IL1β was injected 120 adult male Wistar rats tMCAO induced an increase in brain water content, necrosis, and neutrophilic infiltration in the cortex perfused by the MCA and the DCP and VCP. rIL-1β into the left lateral ventricle immediately after reperfusion markedly enhanced ischemic brain edema formation and infarction size in MCA zone, DCP, and VCP in a dose-dependent manner (p < 0.01). Anti-IL-1β attenuated the post-ischemic increase of brain water content and decreased the infarction size (p < 0.01). The number of neutrophils infiltrating the ischemic area decreased with anti-IL-1β.
Relton et al. [236] 1996 MCAO or sham surgery. Animals were injected subcutaneously with either vehicle or rIL-1Ra at 0, 4, 8, 12, and 18 h after ischemia. In separate experiments, initial treatment was delayed until 30 min, 1 h, or 4 h after ischemia and treatments were repeated until 18 h Male Sprague-Dawley rats rIL-1Ra significantly inhibited infarct size by 46% at 24 h (p < 0.05), cerebral edema formation by 49% at 24 h (p < 0.05). Infarction inhibition by rIL-1Ra was dependent on dose and time of administration.
Pradillo et al. [234] 2012 Lean and Cp rats received placebo or IL-1Ra (25 and 12.5 mg/kg) subcutaneously at reperfusion and 6 h later and allocated to different groups: lean + tMCAO + placebo; lean + tMCAO + IL-1Ra; Cp + tMCAO + placebo; and Cp + tMCAO + IL-1Ra. For the delayed administration study, animals were injected subcutaneously with placebo or IL-1Ra at 3 h of reperfusion and again 3 h later Male, lean and Cp rats IL-1Ra at reperfusion resulted in a 50% reduction of infarct volume as measured by MRI both in lean and Cp compared with placebo-treated animals (p < 0.05). IL-1Ra decreased the number of MMP-9-positive neutrophils when compared with placebo (p < 0.05). In both lean and Cp rats, IL-1Ra largely reduced the microglial activation compared with the placebo-treated groups (p < 0.05). In 16-month-old lean rats, delayed IL-1Ra significantly reduced the number of MMP-9-positive blood vessels and the number of MMP-9-positive neutrophils when compared with the placebo group (p < 0.05).
Statins
Endres et al. [219] 1998 After tMCAO followed by reperfusion, mice were injected subcutaneously with 0.1 mL of activated simvastatin or lovastatin (0.2–20 mg/kg) or a corresponding volume of PBS once daily for 3 or 14 days Not declared In a concentration-dependent manner, simvastatin for 14 days reduced cerebral infarct size by 18, 27 and 46% (p < 0.05) and increased NOS activity (p < 0.05). Simvastatin 20 mg/kg increased basal hemispheric CBF by 31% (p < 0.05). Lovastatin 20 mg/kg daily for 14 days also decreased cerebral infarct size and neurological deficits, even if to a lesser extent than simvastatin.
Kawashima et al. [222] 2003 Two groups, one statin-treated (cerivastatin 2 mg/kg by gavage once daily) and another vehicle-treated Stroke-prone spontaneously hypertensive rats (4 weeks of age) The incidence of stroke and stroke size decreased (p < 0.01). High-dose statin treatment delayed early death and reduced the occurrence of stroke-associated symptoms (p < 0.01) and decreased stroke-associated infiltration of inflammatory cells (p < 0.05). Statin treatment increased eNOS protein levels and eNOS activity (p < 0.05). Superoxide production was reduced in statin-treated rats (p < 0.01).
Amin-Hanjani et al. [211] 2001 Two groups: mevastatin at a dose of 2 or 20 mg/kg daily and a corresponding concentration of vehicle for 7, 14, or 28 days before tMCAO Wild-type male mice and eNOS-deficient male mice Mevastatin increased levels of eNOS mRNA and protein, reduced infarct size, and improved neurological deficits in a dose- and time-dependent manner especially with 14- and 28-day high-dose treatment (26% and 37% infarct reduction, respectively, p < 0.05). Cholesterol levels were reduced only after 28 days of treatment (p < 0.05), but did not correlate with infarct reduction. Baseline absolute cerebral blood flow was 30% higher after 14-day high-dose treatment (p < 0.05).
Prinz et al. [235] 2008 After tMCAO followed by reperfusion, mice were treated with intravenously or intraperitoneally rosuvastatin given up to 6 h after MCAO (0.02–20 mg/kg) Wild-type mice aged 6 to 8 weeks Intravenous rosuvastatin significantly reduced lesion size up to 4 h after MCAO in doses as low as 0.2 mg/kg (p < 0.05). Intraperitoneal administration provided protection only on reperfusion at a dose of 20 mg/kg (p < 0.05). Lesion protection was evident 5 days after brain ischemia and was associated with functional improvements at 2.0 mg/kg dose (p < 0.05). Neuroprotection with intravenous rosuvastatin was achieved with peak plasma concentrations <0.5 ng/mL and was associated with increased levels of phosphorylated Akt kinase and eNOS in the vasculature (p < 0.05).
Asahi et al. [212] 2005 Heterologous blood clots were used to induce MCAO after long-term simvastatin (20 mg/kg), atorvastatin (20 mg/kg) or vehicle treatment subcutaneously Male SV-129 mice and male C57Bl/6 mice In wild-type mice, both simvastatin and atorvastatin reduced ischemic lesions and residual clot after 14 days (p < 0.05). In eNOS knockout mice, atorvastatin reduced the volume of ischemic tissue and improved neurologic outcomes after arterial occlusion (p < 0.05). Both statins did not have protective effects in t-PA knockout mice after embolic focal ischemia, but only in a filament model where focal ischemia was achieved via mechanical occlusion (p < 0.05).
Chen et al. [215] 2003 24 h after MCAO, rats were fed atorvastatin (1, 3 or 8 mg/kg) daily for 7 days. Rats were also treated with simvastatin 1 mg/kg with the same protocol 48 Adult male Wistar rats Rats treated with 1 and 3 mg/kg atorvastatin and 1 mg/kg simvastatin improved functional recovery (p < 0.05). VEGF production within the ischemic boundary area at 14 days after stroke increased in the 1 mg/kg atorvastatin group (p < 0.05) as well as cyclic guanosine monophosphate, angiogenesis, neurogenesis, and synaptophysin levels (p < 0.05).
Sironi et al. [242] 2003 Two groups of rats were treated with vehicle alone or simvastatin for 3 days before MCAO, while other two groups underwent MCAO and were treated with vehicle or simvastatin at 3 and 25 h after the induction of the injury. The brain infarct size was evaluated using MRI Male Sprague-Dawley rats Treatment with simvastatin (20 mg/kg) after MCAO prevented the increase in brain infarct volume occurring at 24 h and induced a 46.6% reduction after 48 h (p < 0.01). The neuroprotective effects of simvastatin were paralleled by an increase in eNOS immunoreactivity, detectable in the brain of simvastatin-treated rats.
Reuter et al. [238] 2015 Cultured hBMECs pretreated with simvastatin and subjected to OGD hBMECs Simvastatin significantly blocked the expression of MMP-2 under OGD (p < 0.004). MMP-9 synthesis rate was low and unaffected by simvastatin treatment, while the gene expression and protein secretion of TIMP-1 and TIMP-2 were both strongly induced (p < 0.001).
Fingolimod (FTY720)
Rolland et al. [239] 2013 Fingolimod was given intraperitoneally at a dose of 1 mg/kg as single dose 1 h after ICH induction or daily administration 1, 24, and 48 h after ICH induction 103 male CD-1 mice and 28 male Sprague-Dawley rats Fingolimod enhanced neurological functions and reduced brain edema at 24 and 72 h following experimental ICH in CD-1 mice (p < 0.05). Fewer lymphocytes were found in blood and brain samples of treated animals (p < 0.05). Fingolimod decreased ICAM-1, IFN-γ, IL-17 levels 72 h after ICH (p < 0.05). Treated Sprague-Dawley rats showed less spatial and motor learning deficits along with significantly reduced brain atrophy and neuronal cell loss within the basal ganglia (p < 0.05).
Campos et al. [214] 2013 3 cohorts: pMCAO not treated with t-PA; tMCAO followed by early (30 min after thrombin) t-PA administration; and tMCAO followed by delayed (3 h after thrombin) t-PA administration. Each of these cohort received fingolimod at different time points C57BL/6 male mice Fingolimod reduced neurological deficits and infarct volume after in situ thromboembolic MCAO (p < 0.05). Combination of fingolimod and t-PA improved neurological outcomes of the thrombolytic therapy and the risk of hemorrhagic transformation associated with delayed administration of t-PA (p < 0.05).
Donepezil
Wang et al. [244] 2014 3 groups: the sham operation group (SO), the model group (MG) and the treatment group (TG). Pathological appearance of the hippocampal CA1 region and calpain I and CDK5/p25 expression were observed on the 4th, 6th and 8th week from I/R surgery 250 3-month old male mice At each postoperative time point, the normal neuron count of the hippocampal CA1 region in the treatment group increased significantly (p < 0.05), whereas calpain I and CDK5/p25 expression, SOD activity and MDA content were significantly lower than those in the model group (p < 0.05).
Min et al. [231] 2012 After transient global ischemia, donepezil (5 mg/kg once a day) was administered intragastrically for 21 days Male Mongolian gerbils Donepezil significantly inhibited delayed neuronal death in the hippocampal CA1 region (p < 0.01). Memory impairment was significantly improved by donepezil treatment (p < 0.05–0.01). Western blot analysis showed that donepezil treatment prevented reductions in p-CaMKII and p-CREB protein levels in the hippocampus (p < 0.01).
Yuan et al. [252] 2011 Cultured cells were exposed to both OGD and electrophysiological experiment HEK293 cells from a human embryonic kidney cell line Donepezil showed to attenuate OGD-induced apoptosis in Kv2.1/HEK293 cells and to inhibit Kv2.1 currents in a dose-dependent manner under normoxic condition (p < 0.01). Donepezil further inhibited Kv2.1 currents after OGD treatment (p < 0.05).
Akasofu et al. [210] 2008 Prolonged opening of sodium channels with veratridine led to depolarization-induced neuronal cell injury, which was prevented by 0.1 µM tetrodotoxin Cortical cell cultures from fetal rats of the Wistar strain Pre-treatment with donepezil (0.1–10 µM) for 1 day significantly decreased cell death and increased cell viability in a concentration-dependent manner (p < 0.05). At 0.1–10 µM, donepezil concentration-dependently decreased the veratridine-induced increase of calcium concentration, whilst at 10 µM it reduced the veratridine-induced increase of sodium concentration (p < 0.05 for both).
Lee et al. [225] 2007 After permanent ligation of bilateral common carotid arteries, rats were administered cilostazol (30 mg/kg/day orally) and donepezil (0.3 mg/kg/day intraperitoneally) Rats Concurrent treatment with cilostazol and donepezil prevented neuropathological alterations in the white matter by activation of phosphorylated CREB and Bcl-2, resulting in improvement of spatial learning memory (p < 0.05).
Citalopram
Espinera et al. [220] 2013 After focal ischemic stroke, citalopram 10 mg/kg was injected intraperitoneally 24 h after stroke and then daily for 7, 14, 21, or 28 days Adult male C57 mice Citalopram had no significant effect on infarct formation or edema 3 days after stroke, but enhanced sensorimotor functional recovery after 14 days (p < 0.05). Citalopram improved neuroblast proliferation and migration (p < 0.01) as well as neurogenesis (p < 0.05) and peri-infarction vessel density (p < 0.05) in the post-ischemic brain.
Kronenberg et al. [223] 2012 Mice were subjected to 30-min MCAO/reperfusion and serial MRI scans; a subset of animals received citalopram from day 7 after MCAO Male 129/SV mice Delayed citalopram reversed the behavioral phenotype blocked the degeneration of dopaminergic midbrain neurons, and attenuated striatal atrophy after 4 months (p < 0.05).
Natalizumab
Becker et al. [213] 2001 Rats underwent 3 h of MCAO followed by 45 h of reperfusion. 2 h after ischemia, one group received anti-α4 integrin antibody intraperitoneally and another an isotype control antibody Male Lewis rats Neurological deficits were less frequent in treated rats at 24 (p < 0.01) and 48 h (p = 0.01) after ischemia. White blood cell count was higher in treated rats (p < 0.01) with a lymphocyte/monocyte predominance. Infarction volume was reduced in treated animals (p = 0.012).
Relton et al. [237] 2001 Rats underwent 1-h MCAO followed by 23-h reperfusion. 24 h before MCAO were injected intravenously with anti-α4 integrin antibody (2.5 mg/kg) or isotype control antibody Male spontaneously hypertensive rats or Sprague-Dawley rats Treated animals showed reduced total infarct volume (p < 0.05–0.01). Moreover, treatment reduced brain myeloperoxidase activity (p < 0.05). No significant difference in white blood cell count was observed. Leukocyte counts were elevated in TA-2-treated rats.
Liesz et al. [226] 2011 24 h before or 3 h after ischemia, mice were administered 300 mg of CD49d-specific monoclonal antibody intraperitoneally after; control animals received rat IgG2b isotype control monoclonal antibody Male mice C57BL/6J aged 10–12 weeks VLA-4 blockade improved outcome after 7 days from MCAO via the inhibition of cerebral leukocyte invasion and neurotoxic cytokine production (p < 0.01). VLA-4 inhibition reduced the post-ischemic VCAM-1 up-regulation (p < 0.01).
Langhauser et al. [224] 2014 24 h before or 3 h after cerebral ischemia (both tMCAO and pMCAO), mice were treated with 300 μg of a monoclonal antibody anti-CD49d Male C57Bl/6 mice VLA-4 blocking reduced T cell and neutrophil invasion after 5 days following MCAO and inhibited the up-regulation of VCAM-1 (p < 0.05). Anti-CD49d antibody could not influence stroke outcome positively, irrespective of the model or the time point investigated.
Neumann et al. [232] 2015 After focal cerebral ischemia was induced by pMCAO, anti-CD49d treatment was administered intravenously LysM-eGFP mice The systemic blockade of VLA-4 resulted in reduction of adherence of neutrophils (p < 0.05) and inhibition of their infiltration (p < 0.01) 24 h after focal ischemia. Moreover, anti-VLA-4 treatment improved neurological outcome and reduced infarct volume at day 3 after stroke (p < 0.05).
Llovera et al. [230] 2015 After cMCAO (for small lesions confined to the cortex) or fMCAO (for lesions in the cortex and subcortical structures) was assessed, anti-CD49d treatment was administered intraperitoneally 3 h after stroke induction 315 male C57BL/6J mice Anti-CD49d treatment reduced infarct volume (p < 0.05) and leukocytes invasion into the ischemic brain (p < 0.001) after 7 days from cMCAO (p < 0.05). After fMCAO, mice had fewer cerebral leukocytes than after cMCAO (p < 0.001), but anti-CD49d treatment did not affect leukocyte invasion after fMCAO.
Cyclosporine A
Uchino et al. [243] 1998 CsA was given intraperitoneally daily for 1 week before and 1 week after forebrain ischemia of 7 or 10 min duration Rats Systemically administered CsA ameliorated the damage to the CA1 sector of the hippocampus due to transient ischemia (p < 0.001).
Cho et al. [217] 2013 Rats underwent MCAO and then randomly treated by intracarotid CsA 10 mg/kg 20 min before MCAO (pre-treatment group); intracarotid CsA 10 mg/kg immediately after reperfusion (post-treatment); and intracarotid saline immediately after reperfusion 27 Sprague-Dawley rats On day 1, a significant reduction of infarct size in the pre-treatment group compared to the post-treatment (p < 0.004) was evaluated. A significant reduction of microglial cell count in the pre-treatment group compared to either saline or post-treatment groups was found (p < 0.001).
Yu et al. [250] 2004 Rats underwent MCAO then were randomly treated with either: low dose CsA, MP, low dose CsA plus MP, high dose CsA, or vehicle Adult Sprague-Dawley rats Animals receiving high dose CsA alone exhibited a minor motor asymmetry and less neurologic deficits 3 days after stroke (p < 0.0001) as well as those receiving low dose CsA and MP treatment but only on day 1 post-stroke (p <0.005). Animals receiving high dose CsA alone exhibited significantly (p < 0.0001).
Yuen et al. [253] 2011 Rats were equally divided into sham control, intraperitoneal physiological saline (at 0.5/24/48 h after stroke), CsA (20 mg/kg at 0.5/24 h intraperitoneally), EPO (5000 IU/kg at 0.5/24/48 h, subcutaneously), combined CsA and EPO after occlusion of distal left internal carotid artery 50 adult-male Sprague-Dawley rats On day 21, improvement in neurological function was found in CsA and EPO group (p < 0.05) and was higher when the combined treatment was administered (p < 0.004). Attenuation of inflammatory response, apoptosis, and oxidative stress was found with combined therapy with CsA and EPO (p < 0.05).
Edaravone
Fujiwara et al. [221] 2016 Before 90-min MCAO followed by reperfusion, rats were randomly assigned to intravenous vehicle or intravenous edaravone 3 mg/kg Male Sprague-Dawley rats Edaravone decreased infarct volume and edema formation and IL-1β and MMP-9 levels 3 h after ischemia levels (p < 0.05). Edaravone was shown to reduce levels of many other pro-inflammatory cytokines.
Yamashita et al. [248] 2015 Thrombolysis was evaluated by using a He-Ne-laser-induced thrombosis model in mesenteric microvessels. 3 experimental groups (placebo, alteplase 0.6 mg/kg, alteplase 0.6 mg/kg + edaravone 10.5 mg/kg) Male Wistar–ST rats In the alteplase group, thrombus volume decreased (p < 0.01) after 20 min. In the alteplase+edaravone group, thrombus volume was more evident (p < 0.001).
Wu et al. [245] 2014 Rats were subjected to tMCAO and then administered edaravone 2.4 mg/kg; a subset of these animals were administered both edaravone 2.4 mg/kg and borneol 0.6 mg/kg Sprague-Dawley rats Edaravone was demonstrated to scavenge free radicals. Edavarone and borneol reduced the infarct area (p < 0.001) and the effect was increased when drugs were administered synergistically (p < 0.001).

IL-1Ra: interleukin-1 receptor antagonist; pMCAO: permanent middle cerebral artery occlusion; CV: cardiovascular; PMN: polymorphonuclear; tMCAO: transient middle cerebral artery occlusion; DCP: dorsal area of caudate putamen; VCP: ventral area of the caudate putamen; rIL-1β: recombinant interleukin-1β; MRI: magnetic resonance imaging; MMP: metalloproteinase; CBF: cerebral blood flow; eNOS: endothelial nitric oxide synthase; t-PA: tissue-type plasminogen activator; VEGF: vascular endothelial growth factor; hBMEC: human brain microvascular endothelial cells; OGD: oxygen glucose deprivation; ICH: intracerebral hemorrhage; ICAM-1: intercellular adhesion molecule-1; IFN-γ: interferon-γ; IL: interleukin; I/R: ischemia/reperfusion; CDK5: cyclin-dependent kinase 5; SOD: superoxide dismutase; MDA: malondialdehyde; CaMKII: calmodulin-dependent protein kinase II; CREB: cyclic adenosine monophosphate responsive element binding protein; Kv channels: voltage-gated potassium channels; VCAM-1: vascular cell adhesion molecule-1; VLA-4: very late antigen-4; LysM–EGFP: lysozyme M promoter driving expression of enhanced green fluorescent protein; cMCAO: coagulation of the distal middle cerebral artery; fMCAO: occlusion of the middle cerebral artery with an endovascular filament; CsA: cyclosporine A; MP: methylprednisolone; EPO: erythropoietin.