losartan-potassium and Infarction--Middle-Cerebral-Artery

losartan-potassium has been researched along with Infarction--Middle-Cerebral-Artery* in 50 studies

Trials

1 trial(s) available for losartan-potassium and Infarction--Middle-Cerebral-Artery

ArticleYear
Recombinant human erythropoietin in the treatment of acute ischemic stroke.
    Stroke, 2009, Volume: 40, Issue:12

    Numerous preclinical findings and a clinical pilot study suggest that recombinant human erythropoietin (EPO) provides neuroprotection that may be beneficial for the treatment of patients with ischemic stroke. Although EPO has been considered to be a safe and well-tolerated drug over 2 decades, recent studies have identified increased thromboembolic complications and/or mortality risks on EPO administration to patients with cancer or chronic kidney disease. Accordingly, the double-blind, placebo-controlled, randomized German Multicenter EPO Stroke Trial (Phase II/III; ClinicalTrials.gov Identifier: NCT00604630) was designed to evaluate efficacy and safety of EPO in stroke.. This clinical trial enrolled 522 patients with acute ischemic stroke in the middle cerebral artery territory (intent-to-treat population) with 460 patients treated as planned (per-protocol population). Within 6 hours of symptom onset, at 24 and 48 hours, EPO was infused intravenously (40,000 IU each). Systemic thrombolysis with recombinant tissue plasminogen activator was allowed and stratified for.. Unexpectedly, a very high number of patients received recombinant tissue plasminogen activator (63.4%). On analysis of total intent-to-treat and per-protocol populations, neither primary outcome Barthel Index on Day 90 (P=0.45) nor any of the other outcome parameters showed favorable effects of EPO. There was an overall death rate of 16.4% (n=42 of 256) in the EPO and 9.0% (n=24 of 266) in the placebo group (OR, 1.98; 95% CI, 1.16 to 3.38; P=0.01) without any particular mechanism of death unexpected after stroke.. Based on analysis of total intent-to-treat and per-protocol populations only, this is a negative trial that also raises safety concerns, particularly in patients receiving systemic thrombolysis.

    Topics: Acute Disease; Adult; Aged; Aged, 80 and over; Brain Ischemia; Double-Blind Method; Drug Administration Schedule; Drug Interactions; Drug-Related Side Effects and Adverse Reactions; Erythropoietin; Female; Humans; Infarction, Middle Cerebral Artery; Injections, Intravenous; Male; Middle Aged; Mortality; Neuroprotective Agents; Patient Selection; Placebo Effect; Recombinant Proteins; Stroke; Tissue Plasminogen Activator; Treatment Outcome; Young Adult

2009

Other Studies

49 other study(ies) available for losartan-potassium and Infarction--Middle-Cerebral-Artery

ArticleYear
Erythropoietin attenuates axonal injury after middle cerebral artery occlusion in mice.
    Neurological research, 2017, Volume: 39, Issue:6

    Erythropoietin (EPO) confers potent neuroprotection against ischemic injury through a variety of mechanisms. However, the protective effect of EPO on axons after cerebral ischemia in adult mice is rarely covered. The purpose of this study was to investigate the potential neuroprotective effects of EPO on axons in mice after cerebral ischemia.. A total of 30 adult male C57 BL/6 mice were treated with EPO (5000 IU/kg) or vehicle after transient middle cerebral artery occlusion (MCAO). The mortality rate of each experimental group was calculated. Neurological function was assessed by Rota-rod test. Frozen sections from each mouse brain at 14 days after reperfusion were used to evaluate the fluorescent intensity of myelin basic protein (MBP) and neurofilament 200 (NF-200). Immunofluorescence staining and Western blotting were used to assess the protein level of β-amyloid precursor protein (β-APP) and glial fibrillary acidic protein (GFAP), a marker of mature astrocytes. The protein levels of the myelin-derived growth inhibitory proteins, neurite growth inhibitor-A (Nogo-A), myelin-associated glycoprotein (MAG) and oligodendrocyte-myelin glycoprotein (OMG) were also examined by Western blot after MCAO.. The survival rate of the vehicle group 14 days after cerebral ischemia-reperfusion was 50%, which increased to 80% after EPO treatment at the start of reperfusion. EPO improved neurobehavioral outcomes at days 3 and 7 after MCAO was compared with the vehicle group (P < 0.05). Furthermore, EPO ameliorated demyelination, demonstrated by upregulation of the MBP/NF-200 ratio. Meanwhile, increased levels of β-APP, GFAP, Nogo-A, and MAG after MCAO were reduced by EPO treatment (P < 0.05).. EPO treatment attenuates axonal injury and improves neurological function after cerebral ischemia in adult mice.

    Topics: Animals; Axons; Disease Models, Animal; Erythropoietin; Infarction, Middle Cerebral Artery; Male; Mice, Inbred C57BL; Neuroprotective Agents; Nogo Proteins

2017
The prolyl 4-hydroxylase inhibitor GSK360A decreases post-stroke brain injury and sensory, motor, and cognitive behavioral deficits.
    PloS one, 2017, Volume: 12, Issue:9

    There is interest in pharmacologic preconditioning for end-organ protection by targeting the HIF system. This can be accomplished by inhibition of prolyl 4-hydroxylase (PHD). GSK360A is an orally active PHD inhibitor that has been previously shown to protect the failing heart. We hypothesized that PHD inhibition can also protect the brain from injuries and resulting behavioral deficits that can occur as a result of surgery. Thus, our goal was to investigate the effect of pre-stroke surgery brain protection using a verified GSK360A PHD inhibition paradigm on post-stroke surgery outcomes. Vehicle or an established protective dose (30 mg/kg, p.o.) of GSK360A was administered to male Sprague-Dawley rats. Initially, GSK360A pharmacokinetics and organ distribution were determined, and then PHD-HIF pharmacodynamic markers were measured (i.e., to validate the pharmacological effects of the GSK360A administration regimen). Results obtained using this validated PHD dose-regimen indicated significant improvement by GSK360A (30mg/kg); administered at 18 and 5 hours prior to transient middle cerebral artery occlusion (stroke). GSK360A exposure and plasma, kidney and brain HIF-PHD pharmacodynamics endpoints (e.g., erythropoietin; EPO and Vascular Endothelial Growth Factor; VEGF) were measured. GSK360A provided rapid exposure in plasma (7734 ng/ml), kidney (45-52% of plasma level) and brain (1-4% of plasma level), and increased kidney EPO mRNA (80-fold) and brain VEGF mRNA (2-fold). We also observed that GSK360A increased plasma EPO (300-fold) and VEGF (2-fold). Further assessments indicated that GSK360A reduced post-stroke surgery neurological deficits (47-64%), cognitive dysfunction (60-75%) and brain infarction (30%) 4 weeks later. Thus, PHD inhibition using GSK360A pretreatment produced long-term post-stroke brain protection and improved behavioral functioning. These data support PHD inhibition, specifically by GSK360A, as a potential strategy for pre-surgical use to reduce brain injury and functional decline due to surgery-related cerebral injury.

    Topics: Administration, Oral; Animals; Behavior, Animal; Brain; Brain Injuries; Cognition Disorders; Erythropoietin; Glycine; Hypoxia-Inducible Factor 1, alpha Subunit; Infarction, Middle Cerebral Artery; Male; Motor Activity; Organ Specificity; Prolyl Hydroxylases; Prolyl-Hydroxylase Inhibitors; Quinolones; Rats, Sprague-Dawley; RNA, Messenger; Sensation; Stroke; Vascular Endothelial Growth Factor A

2017
Delayed erythropoietin therapy improves histological and behavioral outcomes after transient neonatal stroke.
    Neurobiology of disease, 2016, Volume: 93

    Stroke is a major cause of neonatal morbidity, often with delayed diagnosis and with no accepted therapeutic options. The purpose of this study is to investigate the efficacy of delayed initiation of multiple dose erythropoietin (EPO) therapy in improving histological and behavioral outcomes after early transient ischemic stroke.. 32 postnatal day 10 (P10) Sprague-Dawley rats underwent sham surgery or transient middle cerebral artery occlusion (tMCAO) for 3h, resulting in injury involving the striatum and parieto-temporal cortex. EPO (1000U/kg per dose×3 doses) or vehicle was administered intraperitoneally starting one week after tMCAO (at P17, P20, and P23). At four weeks after tMCAO, sensorimotor function was assessed in these four groups (6 vehicle-sham, 6 EPO-sham, 10 vehicle-tMCAO and 10 EPO-tMCAO) with forepaw preference in cylinder rearing trials. Brains were then harvested for hemispheric volume and Western blot analysis.. EPO-tMCAO animals had significant improvement in forepaw symmetry in cylinder rearing trials compared to vehicle-tMCAO animals, and did not differ from sham animals. There was also significant preservation of hemispheric brain volume in EPO-tMCAO compared to vehicle-tMCAO animals. No differences in ongoing cell death at P17 or P24 were noted by spectrin cleavage in either EPO-tMCAO or vehicle-tMCAO groups.. These results suggest that delayed EPO therapy improves both behavioral and histological outcomes at one month following transient neonatal stroke, and may provide a late treatment alternative for early brain injury.

    Topics: Animals; Animals, Newborn; Brain; Brain Injuries; Cell Death; Erythropoietin; Hypoxia-Ischemia, Brain; Infarction, Middle Cerebral Artery; Neurons; Rats, Sprague-Dawley; Stroke

2016
Catalpol stimulates VEGF production via the JAK2/STAT3 pathway to improve angiogenesis in rats' stroke model.
    Journal of ethnopharmacology, 2016, Sep-15, Volume: 191

    Catalpol is the main active component of the radix from Rehmannia glutinosa Libosch, which has pleiotropic protective effects in neurodegenerative diseases, ischemic stroke, metabolic disorders and others. Catalpol has been shown to have neuroprotective, neurorepair, and angiogenesis effects following ischemic brain injury. However, its molecular mechanisms are still poorly understood. In previous studies, the JAK2/STAT3 signaling pathway was found to play a role in neuroprotection and angiogenesis. This study investigated the role of catalpol in stimulating angiogenesis via the JAK2/STAT3 pathway after permanent focal cerebral ischemia (pMCAO).. Rats were subjected to right middle cerebral artery occlusion through electrocoagulation and were treated with catalpol (5mg/kg), AG490 was also used to inhibit STAT3 phosphorylation (pSTAT3).. Following stroke, Catalpol improved the neuroethology deficit, increased the cerebral blood flow (CBF) of infarcted brain and upregulated EPO and EPOR. AG490 suppressed the phosphorylation of signal transducer and activator of transcription 3 (STAT3), ultimately inhibited VEGF mRNA expression, which reduced VEGF protein expression and inhibited stroke-induced angiogenesis. However, Catalpol enhanced stroke-induced STAT3 activation and subsequently restored STAT3 activity through the recovery of STAT3 binding to VEGF. Moreover, Catalpol reversed the effect of AG490 on STAT3 activation and nuclear translocation, restored the transcriptional activity of the VEGF promoter by recruiting STAT3 to the VEGF promoter, improved VEGF mRNA and protein expression, increased angiogenesis, reduced the difference in CBF between the infarcted and intact brain and ameliorated the neuroethology behaviors after stroke.. Catalpol affects neuroprotection and angiogenesis via the JAK2/STAT3 signaling pathway, which is mediated by STAT3 activation and VEGF expression. Catalpol may be used as a potential therapeutic drug for stroke.

    Topics: Angiogenesis Inducing Agents; Animals; Brain; Cerebral Arteries; Cerebrovascular Circulation; Disease Models, Animal; Erythropoietin; Infarction, Middle Cerebral Artery; Iridoid Glucosides; Janus Kinase 2; Male; Neovascularization, Physiologic; Neuroprotective Agents; Phosphorylation; Promoter Regions, Genetic; Protein Binding; Rats, Sprague-Dawley; Receptors, Erythropoietin; RNA, Messenger; Signal Transduction; STAT3 Transcription Factor; Time Factors; Transcriptional Activation; Up-Regulation; Vascular Endothelial Growth Factor A

2016
Delivery of Hypoxia-Inducible Heme Oxygenase-1 Gene for Site-Specific Gene Therapy in the Ischemic Stroke Animal Model.
    Pharmaceutical research, 2016, Volume: 33, Issue:9

    To reduce side effects due to non-specific expression, the heme oxygenase-1 (HO-1) gene under control of a hypoxia-inducible erythropoietin (Epo) enhancer (pEpo-SV-HO-1) was developed for site-specific gene therapy of ischemic stroke.. pEpo-SV-HO-1 was constructed by insertion of the Epo enhancer into pSV-HO-1. Dexamethasone-conjugated polyamidoamine (PAMAM-Dexa) was used as a gene carrier. In vitro transfection assays were performed in the Neuro2A cells. In vivo efficacy of pEpo-SV-HO-1 was evaluated in the transient middle cerebral artery occlusion (MCAO) model.. In vitro transfection assay with the PAMAM-Dexa/pEpo-SV-HO-1 complex showed that pEpo-SV-HO-1 had higher HO-1 gene expression than pSV-HO-1 under hypoxia. In addition, pEpo-SV-HO-1 reduced the level of apoptosis more efficiently than pSV-HO-1 in Neuro2A cells under hypoxia. For in vivo evaluation, the PAMAM-Dexa/pEpo-SV-HO-1 complex was injected into the ischemic brain of the transient MCAO model. pEpo-SV-HO-1 increased HO-1 expression and reduced the number of apoptotic cells in the ischemic brain, compared with the pSV-HO-1 injection group. As a result, the infarct volume was more efficiently decreased by pEpo-SV-HO-1 than by pSV-HO-1.. pEpo-SV-HO-1 induced HO-1 gene expression and therapeutic effect in the ischemic brain. Therefore, pEpo-SV-HO-1 may be useful for site-specific gene therapy of ischemic stroke.

    Topics: Animals; Brain; Cell Line, Tumor; Dexamethasone; Disease Models, Animal; Erythropoietin; Gene Transfer Techniques; Genetic Therapy; Heme Oxygenase-1; Hypoxia; Infarction, Middle Cerebral Artery; Male; Polyamines; Rats; Rats, Sprague-Dawley; Stroke; Transfection

2016
Intra-artery infusion of recombinant human erythropoietin reduces blood-brain barrier disruption in rats following cerebral ischemia and reperfusion.
    The International journal of neuroscience, 2015, Volume: 125, Issue:9

    Intra-artery infusion of recombinant human erythropoietin (rhEPO) has recently been reported to confer neuroprotection against cerebral ischemia-reperfusion injury in animal models; however, the molecular mechanisms are still under investigation. The present study focused on the specific mechanism involved in blood-brain barrier (BBB) disruption.. Thirty-six male and nine female Sprague Dawley rats were subjected to middle cerebral artery (MCA) occlusion to induce focal cerebral ischemia, and administrated rhEPO at a dose of 800 U/kg through MCA infusion at the beginning of reperfusion. Neurobehavioral deficits, brain edema, and infarct volume were evaluated after 2 h of ischemia and 24 h of reperfusion. BBB permeability was assessed by quantifying the extravasation of Evans blue (EB) dye. The expression of tight junction proteins and matrix metalloproteinases (MMPs) (Claudin-5, Occludin, MMP-2, and MMP-9) in microvessels were detected by immunofluorescence and western blot. The activities of MMPs in the cerebral microvessels were determined by gelatin zymography.. Treatment with rhEPO through the MCA strongly alleviated infarct volume, brain edema, and improved neurobehavioral outcomes in male and female rats. In addition, rhEPO remarkably suppressed the EB extravasation induced by brain ischemia. Furthermore, rhEPO prevented degradation of Claudin-5 and Occludin, and reduced the expression and activity of MMP-2 and MMP-9 in isolated brain microvessels.. Treatment with rhEPO through MCA infusion prevented brain edema formation and infarction through inhibition of MMP-mediated BBB disruption in acute ischemic stroke.

    Topics: Animals; Blood-Brain Barrier; Brain Edema; Brain Ischemia; Cerebral Infarction; Erythropoietin; Female; Humans; Infarction, Middle Cerebral Artery; Infusions, Intra-Arterial; Male; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Reperfusion Injury

2015
Erythropoietin delivered via intra-arterial infusion reduces endoplasmic reticulum stress in brain microvessels of rats following cerebral ischemia and reperfusion.
    Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology, 2015, Volume: 10, Issue:1

    Local infusion of low dose erythropoietin (EPO) alleviates cerebral ischemia and reperfusion (I/R) injury in rats; however, the underlying molecular mechanisms are still unclear. The present study investigated the effect of low dose EPO treatment on I/R-induced endoplasmic reticulum (ER) stress in brain tissue and isolated microvessels in rodents. Sprague-Dawley rats were subjected to 2 h ischemia/24 h reperfusion by middle cerebral artery (MCA) occlusion, then administered fluorescein isothiocyanate-labeled EPO via MCA infusion (MCAI) or subcutaneous injection (SI) to compare the efficiency of two modes of delivery. Neurobehavioral deficits and infarct volume, and the expression of ER stress-associated proteins and apoptosis in brain tissue or isolated microvessels, as well as the transcriptional activity of 16 factors involved in ER stress and the unfolded protein response in brain tissue was asscessed. A higher EPO level in cerebrospinal fluid and brain tissue was observed in rats treated with EPO by MCAI (800 IU/kg) than by SI (5000 IU/kg). Moreover, neurobehavioral deficits and infarct volume were reduced in rats treated with EPO by MCAI and salubrinal. EPO suppressed the expression of ER stress signals glucose-regulated protein 78, activating transcription factor (ATF) 6α, and CCAAT enhancer-binding protein homologous protein (CHOP), as well as that of the pro-apoptotic protein caspase-3 in brain microvessels, and decreased the number of CHOP-positive, apoptotic neurons. EPO treatment also reduced the transcriptional activities of CHOP, forkhead box protein O1, and ATF4. These results provide evidence that low dose EPO treatment via MCAI provides neuroprotection following acute ischemic stroke by inhibiting the ER stress response.

    Topics: Animals; Apoptosis Regulatory Proteins; Brain Ischemia; Capillaries; Cerebrovascular Circulation; Endoplasmic Reticulum Stress; Epoetin Alfa; Erythropoietin; Heat-Shock Proteins; Infarction, Middle Cerebral Artery; Infusions, Intra-Arterial; Male; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Reperfusion Injury

2015
Protection of erythropoietin against ischemic neurovascular unit injuries through the effects of connexin43.
    Biochemical and biophysical research communications, 2015, Mar-13, Volume: 458, Issue:3

    Erythropoietin (EPO) has protective effects on many neurological diseases, including cerebral ischemia. Here, we aimed to test EPO's effects on ischemic neurovascular unit (NVU) injuries and examine whether the effects were dependent on connexin43 (Cx43) mediated gap junctional intercellular communication (GJIC). We detected the expression of Cx43 and phosphorylation of Cx43 (p-Cx43) at 1 d, 3 d, and 7 d after middle cerebral artery occlusion (MCAO). Meanwhile, we examined the effects of EPO on NVU injuries including neuronal survival, astrocyte activation and regeneration of endothelial cells as well as whether the effects were Cx43 dependent by using multiple inhibitors. We found EPO highly increased p-Cx43, but not total Cx43 at all chosen times. Importantly, EPO led to neurological and blood-brain barrier functions improvement by associating with promotion of angiogenesis as well as reduction of neuronal death, astrocyte activation and neurotoxic substances levels. Moreover, these effects were significantly weakened by the inhibitors blocking GJIC, Cx43 communicative function, phosphorylation and expression, only Cx43 redistribution inhibitor excluded. Our data suggest the protective effects of EPO on NUV injuries are highly associated with the increase of p-Cx43, which improves GJIC to reduce neurotoxic substances.

    Topics: Animals; Calcium; Cell Communication; Connexin 43; Erythropoietin; Gap Junctions; Gene Expression; Glutamic Acid; Infarction, Middle Cerebral Artery; Male; Neurons; Neuroprotective Agents; Rats, Sprague-Dawley; RNA Interference; RNA, Small Interfering

2015
Astrocyte-mediated ischemic tolerance.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2015, Mar-04, Volume: 35, Issue:9

    Preconditioning (PC) using a preceding sublethal ischemic insult is an attractive strategy for protecting neurons by inducing ischemic tolerance in the brain. Although the underlying molecular mechanisms have been extensively studied, almost all studies have focused on neurons. Here, using a middle cerebral artery occlusion model in mice, we show that astrocytes play an essential role in the induction of brain ischemic tolerance. PC caused activation of glial cells without producing any noticeable brain damage. The spatiotemporal pattern of astrocytic, but not microglial, activation correlated well with that of ischemic tolerance. Interestingly, such activation in astrocytes lasted at least 8 weeks. Importantly, inhibiting astrocytes with fluorocitrate abolished the induction of ischemic tolerance. To investigate the underlying mechanisms, we focused on the P2X7 receptor as a key molecule in astrocyte-mediated ischemic tolerance. P2X7 receptors were dramatically upregulated in activated astrocytes. PC-induced ischemic tolerance was abolished in P2X7 receptor knock-out mice. Moreover, our results suggest that hypoxia-inducible factor-1α, a well known mediator of ischemic tolerance, is involved in P2X7 receptor-mediated ischemic tolerance. Unlike previous reports focusing on neuron-based mechanisms, our results show that astrocytes play indispensable roles in inducing ischemic tolerance, and that upregulation of P2X7 receptors in astrocytes is essential.

    Topics: Animals; Astrocytes; Brain Ischemia; Erythropoietin; Hypoxia-Inducible Factor 1, alpha Subunit; Infarction, Middle Cerebral Artery; Ischemic Attack, Transient; Ischemic Preconditioning; Mice; Mice, Inbred C57BL; Mice, Knockout; Microglia; Receptors, Purinergic P2X7

2015
Targeted delivery of erythropoietin by transcranial focused ultrasound for neuroprotection against ischemia/reperfusion-induced neuronal injury: a long-term and short-term study.
    PloS one, 2014, Volume: 9, Issue:2

    Erythropoietin (EPO) is a neuroprotective agent against cerebral ischemia/reperfusion (I/R)-induced brain injury. However, its crossing of blood-brain barrier is limited. Focused ultrasound (FUS) sonication with microbubbles (MBs) can effectively open blood-brain barrier to boost the vascular permeability. In this study, we investigated the effects of MBs/FUS on extending the therapeutic time window of EPO and its neuroprotective effects in both acute and chronic phases. Male Wistar rats were firstly subjected to two common carotid arteries and right middle cerebral artery occlusion (three vessels occlusion, 3VO) for 50 min, and then the rats were treated with hEPO (human recombinant EPO, 5000 IU/kg) with or without MBs/FUS at 5 h after occlusion/reperfusion. Acute phase investigation (I/R, I/R+MBs/FUS, I/R+hEPO, and I/R+hEPO+MBs/FUS) was performed 24 h after I/R; chronic tests including cylinder test and gait analysis were performed one month after I/R. The experimental results showed that MBs/FUS significantly increased the cerebral content of EPO by bettering vascular permeability. In acute phase, both significant improvement of neurological score and reduction of infarct volume were found in the I/R+hEPO+MBs/FUS group, as compared with I/R and I/R+hEPO groups. In chronic phase, long-term behavioral recovery and neuronal loss in brain cortex after I/R injury was significantly improved in the I/R+hEPO+MBs/FUS group. This study indicates that hEPO administration with MBs/FUS sonication even at 5 h after occlusion/reperfusion can produce a significant neuroprotection.

    Topics: Animals; Behavior, Animal; Blood-Brain Barrier; Brain Ischemia; Drug Delivery Systems; Erythropoietin; Gait; Infarction, Middle Cerebral Artery; Male; Microbubbles; Neurons; Neuroprotective Agents; Rats; Rats, Wistar; Reperfusion Injury; Sound

2014
Neuroprotective effects of erythropoietin pretreatment in a rodent model of transient middle cerebral artery occlusion.
    Journal of neurosurgery, 2014, Volume: 121, Issue:1

    OBJECT.: There is an unmet clinical need to develop neuroprotective agents for neurosurgical and endovascular procedures that require transient cerebral artery occlusion. The aim in this study was to explore the effects of a single dose of recombinant human erythropoietin (rhEPO) before middle cerebral artery (MCA) occlusion in a focal cerebral ischemia/reperfusion model.. Twenty-eight adult male Wistar rats were subjected to right MCA occlusion via the intraluminal thread technique for 60 minutes under continuous cortical perfusion monitoring by laser Doppler flowmetry. Rats were divided into 2 groups: control and treatment. In the treated group, rhEPO (1000 IU/kg intravenously) was administered 10 minutes before the onset of the MCA ischemia. At 24-hour reperfusion, animals were examined for neurological deficits, blood samples were collected, and animals were killed. The following parameters were evaluated: brain infarct volume, ipsilateral hemispheric edema, neuron-specific enolase plasma levels, parenchyma histological features (H & E staining), Fluoro-Jade-positive neurons, p-Akt and total Akt expression by Western blot analysis, and p-Akt-positive nuclei by immunohistochemical investigation.. Infarct volume and Fluoro-Jade staining of degenerating neurons in the infarct area did not vary between groups. The severity of neurological deficit (p < 0.001), amount of brain edema (78% reduction in treatment group, p < 0.001), and neuron-specific enolase plasma levels (p < 0.001) were reduced in the treatment group. Perivascular edema was histologically less marked in the treatment group. No variations in the expression or localization of p-Akt were seen.. Administration of rhEPO before the onset of 60-minute transient MCA ischemia protected the brain from this insult. It is unlikely that rhEPO pretreatment leads to direct neuronal antiapoptotic effects, as supported by the lack of Akt activation, and its benefits are most probably related to an indirect effect on brain edema as a consequence of blood-brain barrier preservation. Although research on EPO derivatives is increasing, rhEPO acts through distinct neuroprotective pathways and its clinical safety profile is well known. Clinically available rhEPO is a potential therapy for prevention of neuronal injury induced by transitory artery occlusion during neurovascular procedures.

    Topics: Animals; Apoptosis; Blood-Brain Barrier; Brain; Erythropoietin; Infarction, Middle Cerebral Artery; Ischemic Attack, Transient; Male; Neuroprotective Agents; Phosphopyruvate Hydratase; Phosphorylation; Proto-Oncogene Proteins c-akt; Rats; Rats, Wistar

2014
Neuroprotective effects of erythropoietin in focal brain ischemia in rats.
    Bulletin of experimental biology and medicine, 2014, Volume: 156, Issue:5

    Neuroprotective effect of erythropoietin administered before ischemia has been previously demonstrated. The efficiency of erythropoietin administration after ischemia was not studied, though in case of success these protocols would be applied in clinical neurology. In our experiments on the model of transitory focal ischemia, erythropoietin was injected intraperitoneally during the early and delayed postischemic period (3 and 12 h). The size of the necrotic zone, neurological deficit, and the severity of brain edema were evaluated in 48 h. Injection of erythropoietin in 3 and 12 h after ischemia significantly reduced the size of necrosis (p = 0.0007 and p=0.0016, respectively), neurological deficit (p=0.0013 and p=0.0062, respectively), and brain edema (p=0.02 and p=0.0186, respectively). Injection of erythropoietin after transitory focal cerebral ischemia produced a pronounced neuroprotective effect.

    Topics: Animals; Brain; Erythropoietin; Infarction, Middle Cerebral Artery; Injections, Intraperitoneal; Male; Neuroprotective Agents; Rats, Wistar

2014
The neuroprotective mechanism of erythropoietin-TAT fusion protein against neurodegeneration from ischemic brain injury.
    CNS & neurological disorders drug targets, 2014, Volume: 13, Issue:8

    To compare the neuroprotection of erythropoietin (EPO) and EPO fusion protein containing transduction domain derived from HIV TAT (EPO-TAT) against ischemic brain injury, inclusive of the side effect, and explore the mechanism underlying the role of EPO-TAT in a transient focal cerebral ischemia model in rats.. Transient focal ischemia was induced by middle cerebral artery occlusion (MCAO) in rats. Rats were treated, respectively, with following regimens: saline, 1000 U/kg EPO, 5000 U/kg EPO, 1000 U/kg EPO-TAT, 1000 U/kg EPOTAT+5 µl of 10 mM LY294002 (or/plus 5 µl of 5 mM PD98059). Neurological deficit scores, infarct volume, and hematologic side effect were assessed at 72 hours after MCAO. Apoptotic cells were determined with TUNEL staining. The expression and localization of phosphorylated AKT (pAKT) and phosphorylated ERK (pERK) were detected with Western blot, immunohistochemistry, and immunofluorescence, respectively.. 1000 U/kg EPO-TAT exhibited a comparable neuroprotection to 5000 U/kg EPO, as evidenced by a comparable attenuation in neurological deficit, infarct volume, and number of apoptotic cells in the rat ischemic cortex after MCAO. The pAKT and pERK levels were significantly elevated solely in neurons of rodents receiving EPO or EPO-TAT treatments, suggesting the concurrent activation of these two pathways. Specific inhibition of either AKT or ERK pathway partially abolished EPO-TAT protection, but exhibited no influence on the activation status of its counterpart, suggesting no cross-modulation between these two protective pathways.. Our study indicates that EPO-TAT at 1000 U/kg displays neuroprotection with no detectable side effects. The mechanism for neuroprotection may be attributable to the simultaneous activation of the AKT and ERK pathways, which preserve neuronal cell viability and attenuate behavioral deficits.

    Topics: Animals; Cell Death; Disease Models, Animal; Enzyme Inhibitors; Erythropoietin; Extracellular Signal-Regulated MAP Kinases; In Situ Nick-End Labeling; Infarction, Middle Cerebral Artery; Male; Neurodegenerative Diseases; Neurologic Examination; Neuroprotective Agents; Oncogene Protein v-akt; Phosphopyruvate Hydratase; Rats; Rats, Sprague-Dawley; Recombinant Fusion Proteins; Statistics, Nonparametric; tat Gene Products, Human Immunodeficiency Virus

2014
Brain ischemic injury in rodents: the protective effect of EPO.
    Methods in molecular biology (Clifton, N.J.), 2013, Volume: 982

    Animal models constitute an indispensable tool to investigate human pathology. Here we describe the procedure to induce permanent and transient cerebral ischemia in the mouse and the rat. The model of transient occlusion of the middle cerebral artery (MCA) is performed by the insertion of an occlusive filament until the origin of the MCA while the permanent occlusion described in the mice is performed by a distal electrocoagulation of the MCA. Those models allow evaluating the efficiency of therapeutic strategy of ischemia from tissular aspect to behavioral and cognitive impairment assessment. They were widely used in the literature to evaluate the efficiency of different drugs including the cytokines and especially erythropoietin (EPO) or its derivatives.

    Topics: Animals; Brain Ischemia; Erythropoietin; Infarction, Middle Cerebral Artery; Mice; Middle Cerebral Artery; Rats

2013
SMND-309, a novel derivative of salvianolic acid B, protects rat brains ischemia and reperfusion injury by targeting the JAK2/STAT3 pathway.
    European journal of pharmacology, 2013, Aug-15, Volume: 714, Issue:1-3

    SMND-309 is a novel derivative of salvianolic acid B, and has shown protective effects against rat cortical neuron damage in vitro and in vivo. However the molecular mechanisms through which SMND-309 affords this protection are unclear. The present study aimed to investigate the mechanisms associated with the protective activities of SMND-309 in a cerebral ischemia and reperfusion injury rat model. In this study, we used AG490, a specific inhibitor of the signaling pathway involving the Janus Kinase 2 (JAK2)/Signal Transducers and Activators of Transcription 3 (STAT3) signaling molecules and suramin, a potent inhibitor of vascular endothelial growth factor (VEGF), to investigate the mechanisms of SMND-309. The cerebral ischemia and reperfusion injury model was induced by performing middle cerebral artery occlusion (MCAO) in the rats. SMND-309 mitigated the effects of ischemia and reperfusion injury on brain by decreasing the infract volume, improving neurological function, increasing the survival of neurons and promoting angiogenesis by increasing the levels of erythropoietin (EPO), erythropoietin receptor (EPOR), phosphorylated JAK2 (P-JAK2), phosphorylated STAT3 (P-STAT3), VEGF and VEGF receptor 2 (Flk-1) in the brain. Our results suggest that SMND-309 provides significant neuroprotective effects against cerebral ischemia and reperfusion injury. The mechanisms of this protection may be attributed to the increased VEGF expression occurring from the JAK2/STAT3 pathway, activated by the increased EPO/EPOR expression in the brain.

    Topics: Animals; Axons; Benzofurans; Brain; Brain Ischemia; Caffeic Acids; Cerebral Infarction; Dendrites; Erythropoietin; Gene Expression Regulation; Infarction, Middle Cerebral Artery; Janus Kinase 2; Male; Neovascularization, Physiologic; Neuroprotective Agents; Phosphoproteins; Platelet Endothelial Cell Adhesion Molecule-1; Rats; Rats, Sprague-Dawley; Receptors, Erythropoietin; Recovery of Function; Reperfusion Injury; Signal Transduction; STAT3 Transcription Factor; Survival Analysis; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factor Receptor-2; Water

2013
Effects of erythropoietin on STAT1 and STAT3 levels following cerebral ischemia-reperfusion in rats.
    The International journal of neuroscience, 2013, Volume: 123, Issue:10

    Cerebral ischemia-reperfusion injury can activate signal transducers and activators of transcription (STAT). STAT1 initiates neuronal apoptosis following cerebral ischemia-reperfusion, while STAT3 is neuroprotective. Erythropoietin (EPO) promotes regeneration through STAT3 and facilitates neuronal survival following ischemia. However, there are few reports on the effects of EPO on phosphorylated STAT1 (P-STAT1) level following cerebral ischemia-reperfusion in rats, and there is no evidence on the simultaneous observation of the four kinds of protein:STAT1, P-STAT1, STAT3, and P-STAT3.. We established a rat focal cerebral ischemia-reperfusion injury model, and used Western blot and immunohistochemical staining to assess the levels of STAT1 and STAT3 expression, and TdT-mediated dUTP-biotin nick end-labeling (TUNEL) was carried out to observe the number of apoptotic cells with or without EPO treatment.. Our findings show that EPO treatment had no significant effect on STAT1 and STAT3 expression, but P-STAT1 and P-STAT3 were slightly decreased and significantly increased, respectively, after EPO treatment. Neurologic deficits, the infarct volume, and the number of apoptotic cells were significantly decreased after EPO treatment.. The results suggest that EPO exerts a neuroprotective effect by influencing STAT3 and STAT1 expression in the area injured by cerebral ischemia-reperfusion.

    Topics: Animals; Apoptosis; Blotting, Western; Epoetin Alfa; Erythropoietin; Hematinics; Immunohistochemistry; In Situ Nick-End Labeling; Infarction, Middle Cerebral Artery; Male; Neurologic Examination; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Reperfusion Injury; STAT1 Transcription Factor; STAT3 Transcription Factor

2013
Therapeutic benefit of a combined strategy using erythropoietin and endothelial progenitor cells after transient focal cerebral ischemia in rats.
    Neurological research, 2013, Volume: 35, Issue:9

    Many studies have demonstrated beneficial effects of either erythropoietin (EPO) or endothelial progenitor cell (EPC) treatment in cerebral ischemia. To improve post-ischemic tissue repair, we investigated the effect of systemic administration of endothelial colony-forming cells (ECFCs), considered as relevant endothelial progenitors due to their specific vasculogenic activity, in the presence or absence of EPO, on functional recovery, apoptosis, angiogenesis, and neurogenesis in a transient focal cerebral ischemia model in the adult rat.. Experimental study.. The rats were divided into four groups 24 hours after ischemia,, namely control, ECFCs, EPO, and ECFCs+EPO, and received a single intravenous injection of ECFCs (5 × 10(6) cells) and/or intraperitoneal administration of EPO (2500 UI/kg per day for 3 days).. Infarct volume, functional recovery, apoptosis, angiogenesis, and neurogenesis were assessed at different time points after ischemia.. The combination of EPO and ECFCs was the only treatment that completely restored neurological function. The ECFCs+EPO treatment was also the most effective to decrease apoptosis and to increase angiogenesis and neurogenesis in the ischemic hemisphere compared to controls and to groups receiving ECFCs or EPO alone.. These results suggest that EPO could act in a synergistic way with ECFCs to potentiate their therapeutic benefits.

    Topics: Animals; Apoptosis; Brain; Brain-Derived Neurotrophic Factor; Combined Modality Therapy; Endothelial Cells; Erythropoietin; Infarction, Middle Cerebral Artery; Ischemic Attack, Transient; Male; Neovascularization, Physiologic; Neurogenesis; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recovery of Function; Stem Cell Transplantation; Stem Cells; Vascular Endothelial Growth Factor A

2013
Erythropoietin increases neurogenesis and oligodendrogliosis of subventricular zone precursor cells after neonatal stroke.
    Stroke, 2013, Volume: 44, Issue:3

    Stroke is a common cause of neonatal brain injury. The subventricular zone is a lifelong source of newly generated cells in rodents, and erythropoietin (EPO) treatment has shown benefit in different animal models of brain injury. The purpose of this study is to investigate the specific role of exogenous EPO on subventricular zone progenitor cell populations in response to neonatal stroke.. Intraventricular injections of green fluorescent protein (GFP)-expressing lentivirus to label subventricular zone precursor cells were made in postnatal day 1 (P1) Long-Evans rats, which then underwent transient middle cerebral artery occlusion on P7. Middle cerebral artery occlusion and sham rats were treated with either vehicle or EPO (1000 U/kg) at reperfusion, 24 hours, and 7 days later. The density of double-labeled DCx+/GFP+, NeuN+/GFP+, O4+/GFP+, GFAP+/GFP+, as well as single-labeled GFP+ and Ki67+ cells, was calculated to determine cell fate outcome in the striatum at 72 hours and 2 weeks after stroke.. There was a significant increase in DCx+/GFP+ and NeuN+/GFP+ neurons and O4+/GFP+ oligodendrocyte precursors, with decreased GFAP+/GFP+ astrocytes at both time points in EPO-middle cerebral artery occlusion animals. There was also a significant increase in GFP+ cells and Ki67+ proliferating cells in EPO compared with vehicle-middle cerebral artery occlusion animals.. These data suggest that subventricular zone neural progenitor cells proliferate and migrate to the site of injury after neonatal stroke and multiple doses of EPO, with a shift in cell fate toward neurogenesis and oligodendrogliosis at both early and late time points. The contribution of local cell proliferation and neurogenesis remains to be determined.

    Topics: Animals; Animals, Newborn; Basal Ganglia; Cell Differentiation; Cell Movement; Cell Proliferation; Dose-Response Relationship, Drug; Doublecortin Protein; Erythropoietin; Green Fluorescent Proteins; Infarction, Middle Cerebral Artery; Models, Animal; Neurogenesis; Oligodendroglia; Rats; Rats, Long-Evans; Stroke; Time Factors

2013
A single injection of liposomal asialo-erythropoietin improves motor function deficit caused by cerebral ischemia/reperfusion.
    International journal of pharmaceutics, 2012, Dec-15, Volume: 439, Issue:1-2

    Modification of the liposomal surface with a targeting molecule is a promising approach for the targeted delivery of therapeutics. Asialo-erythropoietin (AEPO) is a potent tool for targeting an ischemic region by binding to the EPO receptors on neuronal cells. Additionally, it shows a strong cytoprotective effect against programed cell death. Hence, AEPO-modified liposomes appear likely to have both a neuronal-targeting character and a neuroprotective effect on cerebral ischemic injury. In this study, we assessed the targeting ability of AEPO-modified PEGylated liposomes (AEPO-liposomes) to ischemic region and their improvement effect on neurological deficits induced by ischemia/reperfusion (I/R) in transient middle cerebral artery occlusion (t-MCAO) rats. Immunohistological analysis showed that the AEPO-liposomes given immediately after reperfusion extravasated into the ischemic region and attached strongly to neuronal cells. Also, neuronal nuclei (NeuN) staining was clearly visible only in the AEPO-liposome-treated group, suggesting that AEPO-liposomes protected neuronal cells from ischemia/reperfusion-induced damage. Moreover, a single administration of low-dose AEPO-liposomes significantly improved the neurological deficit compared to vehicle and free-AEPO treatment at 7 days after injection. In conclusion, AEPO-liposomes have clear potential as a neuroprotectant after stroke and as a DDS device targeting ischemic regions.

    Topics: Animals; Asialoglycoproteins; Erythropoietin; Infarction, Middle Cerebral Artery; Liposomes; Male; Motor Activity; Neurons; Neuroprotective Agents; Polyethylene Glycols; Rats; Rats, Wistar; Reperfusion Injury

2012
The mitochondria-targeted antioxidants and remote kidney preconditioning ameliorate brain damage through kidney-to-brain cross-talk.
    PloS one, 2012, Volume: 7, Issue:12

    Many ischemia-induced neurological pathologies including stroke are associated with high oxidative stress. Mitochondria-targeted antioxidants could rescue the ischemic organ by providing specific delivery of antioxidant molecules to the mitochondrion, which potentially suffers from oxidative stress more than non-mitochondrial cellular compartments. Besides direct antioxidative activity, these compounds are believed to activate numerous protective pathways. Endogenous anti-ischemic defense may involve the very powerful neuroprotective agent erythropoietin, which is mainly produced by the kidney in a redox-dependent manner, indicating an important role of the kidney in regulation of brain ischemic damage. The goal of this study is to track the relations between the kidney and the brain in terms of the amplification of defense mechanisms during SkQR1 treatment and remote renal preconditioning and provide evidence that the kidney can generate signals inducing a tolerance to oxidative stress-associated brain pathologies.. We used the cationic plastoquinone derivative, SkQR1, as a mitochondria-targeted antioxidant to alleviate the deleterious consequences of stroke. A single injection of SkQR1 before cerebral ischemia in a dose-dependent manner reduces infarction and improves functional recovery. Concomitantly, an increase in the levels of erythropoietin in urine and phosphorylated glycogen synthase kinase-3β (GSK-3β) in the brain was detected 24 h after SkQR1 injection. However, protective effects of SkQR1 were not observed in rats with bilateral nephrectomy and in those treated with the nephrotoxic antibiotic gentamicin, indicating the protective role of humoral factor(s) which are released from functional kidneys. Renal preconditioning also induced brain protection in rats accompanied by an increased erythropoietin level in urine and kidney tissue and P-GSK-3β in brain. Co-cultivation of SkQR1-treated kidney cells with cortical neurons resulted in enchanced phosphorylation of GSK-3β in neuronal cells.. The results indicate that renal preconditioning and SkQR1-induced brain protection may be mediated through the release of EPO from the kidney.

    Topics: Animals; Antioxidants; Brain; Epithelial Cells; Erythropoietin; Gentamicins; Immunoassay; Infarction, Middle Cerebral Artery; Ischemia; Ischemic Preconditioning; Kidney; Kidney Tubules; Male; Mitochondria; Models, Biological; Oxidation-Reduction; Oxidative Stress; Plastoquinone; Rats; Rhodamines

2012
Neuroprotection in stroke in the mouse with intravenous erythropoietin-Trojan horse fusion protein.
    Brain research, 2011, Jan-19, Volume: 1369

    Erythropoietin (EPO) is a potential new treatment for acute stroke. However, EPO does not cross the blood-brain barrier (BBB). EPO has been re-engineered as an IgG-EPO fusion protein, where EPO is fused to the heavy chain of a chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), which is designated the cTfRMAb-EPO fusion protein. The re-engineered EPO is able to penetrate the BBB following intravenous (IV) administration owing to transport on the BBB TfR. In the present study, the neuroprotective properties of EPO alone and the cTfRMAb-EPO fusion protein following IV injection were investigated in a permanent middle cerebral artery occlusion (MCAO) model in the adult mouse. Following MCAO, mice were treated IV with low (1000 U/kg) and high (10,000 U/kg) doses of recombinant EPO, or with low (0.05 mg/kg) or high (1.0 mg/kg) doses of the cTfRMAb-EPO fusion protein. Hemispheric stroke volume and neural deficit scores were quantitated 24h after MCAO. There was no reduction in stroke volume or neural deficit following the IV administration of either dose of EPO or the low dose of cTfRMAb-EPO fusion protein. However, after treatment with the 1.0 mg/kg dose of the cTfRMAb-EPO fusion protein, the hemispheric stroke volume was reduced 81% and the neural deficit was reduced 78%. These studies demonstrate high degrees of neuroprotection in stroke with EPO when the neurotrophin is re-engineered as an IgG-EPO fusion protein to enable transport across the BBB following IV administration.

    Topics: Animals; Antibodies, Monoclonal; Erythropoietin; Immunoglobulin Heavy Chains; Infarction, Middle Cerebral Artery; Injections, Intravenous; Male; Mice; Mice, Inbred C57BL; Neuroprotective Agents; Receptors, Transferrin; Recombinant Fusion Proteins

2011
Neuroprotection by local intra-arterial infusion of erythropoietin after focal cerebral ischemia in rats.
    Neurological research, 2011, Volume: 33, Issue:5

    The neuroprotective effect of erythropoietin has been demonstrated by ischemia and reperfusion models in adult and neonatal rodents. However, administration of high-dose erythropoietin has potential complications. The goal of this study was to determine whether local infusion of low dose erythropoietin offers neuroprotective effects after ischemia and reperfusion injury.. Adult male Sprague-Dawley rats subject to middle cerebral artery occlusion were randomly divided into three groups: (1) sham group: the rats received the same procedure as the other two groups except that no suture was inserted; (2) vehicle group: intra-artery local infusion of saline was administered via middle cerebral artery after reperfusion; and (3) treatment group: 50 U/kg intra-artery local infusion of erythropoietin was administered via middle cerebral artery after reperfusion. Neurological deficit scores and infarct volume (determined by hematoxylin-eosin staining) were evaluated 48 hours after reperfusion. Apoptosis was measured through terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The expression of vascular endothelial growth factor and phosphorylated extracellular signal-regulated kinase were investigated by immunohistochemistry method.. The results show that intra-artery local infusion of erythropoietin, via the middle cerebral artery, significantly reduced neurological deficit scores, foot fault number, and the infarct volume at 48 hours after reperfusion. Significant reductions were also found in the number of positive cells stained by TUNEL assay within the ischemic core and penumbra. Furthermore, local infusion of erythropoietin increased the expression of phosphorylated extracellular signal-regulated kinase and vascular endothelial growth factor.. Local infusion of low-dose erythropoietin via the middle cerebral artery is shown to be neuroprotective against cerebral ischemia and reperfusion injury. The mechanism of neuroprotection may be associated with the increased expression of phosphorylated extracellular signal-regulated kinase and vascular endothelial growth factor.

    Topics: Animals; Brain Ischemia; Disease Models, Animal; Erythropoietin; Infarction, Middle Cerebral Artery; Infusions, Intra-Arterial; Male; Neuroprotective Agents; Random Allocation; Rats; Rats, Sprague-Dawley; Reperfusion Injury

2011
Effect of continuous infusion of asialoerythropoietin on short-term changes in infarct volume, penumbra apoptosis and behaviour following middle cerebral artery occlusion in rats.
    Clinical and experimental pharmacology & physiology, 2010, Volume: 37, Issue:2

    1. Asialoerythropoietin (aEPO), a derivative of cytokine erythropoietin, has been shown to have neuroprotective effects without haematological complications when administered in single or repeated doses. The present study examines our hypothesis that aEPO may provide neuroprotection against programmed apoptotic cell death when administered in a continuous low dose. 2. Focal cerebral ischaemia was introduced by occlusion of the middle cerebral artery using a surgically placed intraluminal filament in young male Sprague Dawley rats (9 weeks old). After 90 min ischaemia, reperfusion was established by filament removal. Both study and control groups had implanted osmotic minipumps through which they received either aEPO (1 microL/h; 20 microg/kg per 24 h) or normal saline (1 microL/h) for 4 days. On Day 4, infarct volume, the number of apoptotic cells and concentrations of activated caspase 3 and 9 were evaluated in the penumbra region. 3. Asialoerythropoietin was detected in the cerebrospinal fluid of the study group, whereas none was detected in the control group. Although there were no significant changes in haematocrit levels or behaviour scores (on Days 1 and 4) between the study and control groups, aEPO administration significantly reduced infarct volume in the study group compared with the control group (168 +/- 19 vs 249 +/- 28 mm(3), respectively; P < 0.05). 4. The number of terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end-labelling (TUNEL)-positive cells and the concentration of activated caspase 3 and 9 in the penumbra region were significantly lower in the study group compared with the control group. 5. In conclusion, our data suggest that aEPO provides a short-term, possibly histological, protection in young adult male rats when administered immediately after reperfusion.

    Topics: Animals; Apoptosis; Asialoglycoproteins; Brain Ischemia; Caspase 3; Caspase 9; Cerebral Infarction; Erythropoietin; Infarction, Middle Cerebral Artery; Male; Neuroprotective Agents; Rats; Rats, Sprague-Dawley

2010
The neuroprotective effect of erythropoietin-transduced human mesenchymal stromal cells in an animal model of ischemic stroke.
    Brain research, 2010, Sep-24, Volume: 1353

    Erythropoietin (EPO) exhibits diverse cellular functions, including neurotrophic, anti-oxidant, anti-apoptotic, and anti-inflammatory effects in non-hematopoietic tissues. This study evaluated whether bone marrow mesenchymal stromal cells (MSCs) transduced with the EPO gene (EPO-MSCs) promoted neural cell survival and improved neurological deficits caused by ischemic stroke. EPO-MSCs stably produced high levels of EPO (10IU/ml) without any alteration of their mesenchymal phenotype. Both EPO transduction and treatment with 10 international units (IU) of recombinant human EPO (rhEPO) provided protection from H(2)O(2)-induced oxidative injury in human bone marrow mesenchymal stromal cells and in SH-SY5Y cells. EPO-MSCs were more protected than were MSCs treated with 10IU rhEPO (10U-MSCs). We also found that the expression of the neurotrophic factors BDNF, PD-ECGF, HGF, SDF-1alpha, and TGF-1beta increased in EPO-MSCs, while only BDNF and TGF-1beta increased in 10U-MSCs. Implantation of EPO-MSCs in an animal model of ischemic stroke significantly improved neurological function and decreased infarct volumes without affecting hematocrit level. An evaluation of the brain tissue 21days after implantation showed that EPO and phosphorylated Akt (a downstream mediator of EPO) increased only in brains implanted with EPO-MSCs. Transduction of the EPO gene into MSCs induced secretion of EPO and various trophic factors that may provide excellent neuroprotective effects in both in vitro and in vivo models of ischemic stroke.

    Topics: Animals; Antigens, CD; Brain Infarction; Caspase 3; Cell Death; Cell Line; Disease Models, Animal; Enzyme-Linked Immunosorbent Assay; Erythropoietin; Flow Cytometry; Gene Expression Regulation; Green Fluorescent Proteins; Humans; Hydrogen Peroxide; Infarction, Middle Cerebral Artery; Lentivirus; Magnetic Resonance Imaging; Mesenchymal Stem Cell Transplantation; Mesenchymal Stem Cells; Nerve Growth Factors; Nervous System Diseases; Rats; Statistics, Nonparametric; Tetrazolium Salts; Thiazoles; Transduction, Genetic

2010
Catalpol increases brain angiogenesis and up-regulates VEGF and EPO in the rat after permanent middle cerebral artery occlusion.
    International journal of biological sciences, 2010, Aug-20, Volume: 6, Issue:5

    To investigate the role and mechanism of catalpol in brain angiogenesis in a rat model of stroke, the effect of catalpol (5 mg/kg; i.p) or vehicle administered 24 hours after permanent middle cerebral artery occlusion (pMCAO) on behavior, angiogenesis, ultra-structural integrity of brain capillary endothelial cells, and expression of EPO and VEGF were assessed. Repeated treatments with Catalpol reduced neurological deficits and significantly improved angiogenesis, while significantly increasing brain levels of EPO and VEGF without worsening BBB edema. These results suggested that catalpol might contribute to infarcted-brain angiogenesis and ameliorate the edema of brain capillary endothelial cells (BCECs) by upregulating VEGF and EPO coordinately.

    Topics: Animals; Blotting, Western; Endothelial Cells; Erythropoietin; Heart; Immunohistochemistry; Infarction, Middle Cerebral Artery; Iridoid Glucosides; Male; Myocardium; Neovascularization, Physiologic; Psychomotor Performance; Rats; Rats, Sprague-Dawley; Up-Regulation; Vascular Endothelial Growth Factor A

2010
Neuroprotection in experimental stroke in the rat with an IgG-erythropoietin fusion protein.
    Brain research, 2010, Nov-11, Volume: 1360

    Erythropoietin (EPO) is a potent neuroprotective agent that could be developed as a new treatment for stroke. However, the blood-brain barrier (BBB) is intact in the early hours after stroke when neuroprotection is still possible, and EPO does not cross the intact BBB. To enable BBB transport, human EPO was re-engineered as an IgG-EPO fusion protein, wherein the IgG part is a monoclonal antibody (MAb) against the human insulin receptor (HIR). The HIRMAb acts as a BBB molecular Trojan horse to ferry the fused EPO across the BBB via transport on the BBB insulin receptor. The HIRMAb part of the HIRMAb-EPO fusion protein does not recognize the rat insulin receptor. However, the EPO part of the fusion protein does recognize the rat EPO receptor. Therefore, the neuroprotective properties of the HIRMAb-EPO fusion protein were investigated with a permanent middle cerebral artery occlusion model in the rat. The HIRMAb-EPO fusion protein was injected into the ipsilateral brain under stereotaxic guidance. High doses of the HIRMAb-EPO fusion protein (61pmol) completely eliminated both cortical and sub-cortical infarction. Lower doses of the fusion protein (4.5pmol) eliminated the cortical infarct with no significant effect on sub-cortical infarct. The neurologic deficit was reduced by 35% and 90%, respectively, by the 4.5 and 61pmol doses of the HIRMAb-EPO fusion protein. In conclusion, these studies demonstrate the biological activity of the HIRMAb-EPO fusion protein in the brain in vivo, and that EPO retains neuroprotective properties following fusion to the HIRMAb BBB Trojan horse.

    Topics: Animals; Cerebral Cortex; Dose-Response Relationship, Drug; Erythropoietin; Functional Laterality; Immunoglobulin G; Infarction, Middle Cerebral Artery; Male; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recombinant Fusion Proteins; Recombinant Proteins; Stereotaxic Techniques; Stroke

2010
MRI identification of white matter reorganization enhanced by erythropoietin treatment in a rat model of focal ischemia.
    Stroke, 2009, Volume: 40, Issue:3

    The objectives of the present study were to: (1) noninvasively identify white matter reorganization and monitor its progress within 6 weeks after the onset of stroke; and (2) quantitatively investigate the effect of recombinant human erythropoietin treatment on this structural change using in vivo measurement of diffusion anisotropy.. Male Wistar rats were subjected to middle cerebral artery occlusion and treated with recombinant human erythropoietin intraperitoneally at a dose of 5000 U/kg of body weight (n=11) or the same volume of saline (n=7) daily for 7 days starting 24 hours after middle cerebral artery occlusion. MRI measurements of T2- and diffusion-weighted images and cerebral blood flow were performed and neurological severity score was assessed at 1 day and weekly for 6 weeks after middle cerebral artery occlusion. Luxol fast blue and Bielschowsky staining were used to demonstrate myelin and axons, respectively.. White matter reorganization occurred along the ischemic lesion boundary after stroke. The region of white matter reorganization seen on the tissue slice coincided with the elevated area on the fractional anisotropy map, which can be accurately identified. The increase in elevated fractional anisotropy pixels corresponded with progress of white matter reorganization and was associated with improvement of neurological function. Treatment with recombinant human erythropoietin after stroke significantly enhanced white matter reorganization, restored local cerebral blood flow, and expedited functional recovery.. White matter reorganization can be detected by fractional anisotropy. Elevated fractional anisotropy pixels may be a good MRI index to stage white matter remodeling and predict functional outcome.

    Topics: Animals; Anisotropy; Axons; Brain; Diffusion Magnetic Resonance Imaging; Erythropoietin; Histocytochemistry; Humans; Image Processing, Computer-Assisted; Infarction, Middle Cerebral Artery; Ischemic Attack, Transient; Male; Myelin Sheath; Rats; Rats, Wistar; Recombinant Proteins; Recovery of Function

2009
Intranasal delivery of erythropoietin plus insulin-like growth factor-I for acute neuroprotection in stroke. Laboratory investigation.
    Journal of neurosurgery, 2009, Volume: 111, Issue:1

    Individually, the cytokines erythropoietin (EPO) and insulin-like growth factor-I (IGF-I) have both been shown to reduce neuronal damage significantly in rodent models of cerebral ischemia. The authors have previously shown that EPO and IGF-I, when administered together, provide acute and prolonged neuroprotection in cerebrocortical cultures against N-methyl-D-aspartate-induced apoptosis. The aim of this study was to determine whether intranasally applied EPO plus IGF-I can provide acute neuroprotection in an animal stroke model and to show that intranasal administration is more efficient at delivering EPO plus IGF-I to the brain when compared with intravenous, subcutaneous, or intraperitoneal administration.. The EPO and IGF-I were administered intranasally to mice that underwent transient middle cerebral artery occlusion (MCAO). Stroke volumes were measured after 1 hour of MCAO and 24 hours of reperfusion. To evaluate the long-term effects of this treatment, behavioral outcomes were assessed at 3, 30, 60, and 90 days following MCAO. Radiography and liquid scintillation were used to visualize and quantify the uptake of radiolabeled 125I-EPO and 125I-IGF-I into the mouse brain after intranasal, intravenous, subcutaneous, or intraperitoneal administration.. Intranasal administration of EPO plus IGF-I reduced stroke volumes within 24 hours and improved neurological function in mice up to 90 days after MCAO. The 125I-EPO and 125I-IGF-I were found in the brain within 20 minutes after intranasal administration and accumulated within the injured areas of the brain. In addition, intranasal administration delivered significantly higher levels of the applied 125I-EPO and 125I-IGF-I to the brain compared with intravenous, subcutaneous, or intraperitoneal administration.. The data demonstrate that intranasal EPO plus IGF-I penetrates into the brain more efficiently than other drug delivery methods and could potentially provide a fast and efficient treatment to prevent chronic effects of stroke.

    Topics: Acute Disease; Administration, Intranasal; Animals; Disease Models, Animal; Drug Delivery Systems; Drug Therapy, Combination; Erythropoietin; Infarction, Middle Cerebral Artery; Insulin-Like Growth Factor I; Iodine Radioisotopes; Male; Mice; Mice, Inbred C57BL; Neuroprotective Agents

2009
A novel neurotrophic therapeutic strategy for experimental stroke.
    Brain research, 2009, Jul-14, Volume: 1280

    Human chorionic gonadotropin (hCG) promotes proliferation of endogenous neural stem cells, and erythropoietin (EPO) promotes differentiation of these cells into neural stem cells. The current study examined effects of sequential administration of these two compounds, initiated 24 h after stroke. At that time, rats were randomized into four treatment groups: hCG+EPO (3 IM doses hCG over 5 days, followed by 3 IV doses EPO over 3 days), hCG+Saline using the same schedule, Saline+EPO using the same schedule, or neither drug (Saline+Saline). The primary endpoint was the composite neurological score, measured 11 times, from 1 h until 12 weeks post-insult. The neurological score was different across treatment groups (p<0.03). Pairwise testing of groups found that the hCG+EPO group had significantly better behavior at 6/10 post-stroke time points as compared to Saline+Saline. The differences observed when comparing the two-drug group with placebo were less apparent when comparing either of the one-drug groups with placebo. The two one-drug treatment arms did not significantly differ at any time point. Treatment with hCG+EPO significantly reduced total lesion volume by 82-89% compared to the other three treatment groups. The current therapeutic strategy improved behavioral outcome and reduced lesion volume with a time window of 24 h after the onset of stroke. The results from these experiments provide new insight into the effects of these two growth factors on stroke in rats, and could suggest a potential for translation into human stroke studies.

    Topics: Analysis of Variance; Animals; Brain; Chorionic Gonadotropin; Erythropoietin; Humans; Infarction, Middle Cerebral Artery; Male; Neuroprotective Agents; Random Allocation; Rats; Rats, Long-Evans; Severity of Illness Index; Time Factors; Treatment Outcome

2009
Delayed hypoxic postconditioning protects against cerebral ischemia in the mouse.
    Stroke, 2009, Volume: 40, Issue:10

    Inspired from preconditioning studies, ischemic postconditioning, consisting of the application of intermittent interruptions of blood flow shortly after reperfusion, has been described in cardiac ischemia and recently in stroke. It is well known that ischemic tolerance can be achieved in the brain not only by ischemic preconditioning, but also by hypoxic preconditioning. However, the existence of hypoxic postconditioning has never been reported in cerebral ischemia.. Adult mice subjected to transient middle cerebral artery occlusion underwent chronic intermittent hypoxia starting either 1 or 5 days after ischemia and brain damage was assessed by T2-weighted MRI at 43 days. In addition, we investigated the potential neuroprotective effect of hypoxia applied after oxygen glucose deprivation in primary neuronal cultures.. The present study shows for the first time that a late application of hypoxia (5 days) after ischemia reduced delayed thalamic atrophy. Furthermore, hypoxia performed 14 hours after oxygen glucose deprivation induced neuroprotection in primary neuronal cultures. We found that hypoxia-inducible factor-1alpha expression as well as those of its target genes erythropoietin and adrenomedullin is increased by hypoxic postconditioning. Further studies with pharmacological inhibitors or recombinant proteins for erythropoietin and adrenomedullin revealed that these molecules participate in this hypoxia postconditioning-induced neuroprotection.. Altogether, this study demonstrates for the first time the existence of a delayed hypoxic postconditioning in cerebral ischemia and in vitro studies highlight hypoxia-inducible factor-1alpha and its target genes, erythropoietin and adrenomedullin, as potential effectors of postconditioning.

    Topics: Adrenomedullin; Animals; Atrophy; Brain; Cells, Cultured; Cytoprotection; Disease Models, Animal; Energy Metabolism; Erythropoietin; Hypoxia-Inducible Factor 1, alpha Subunit; Hypoxia-Ischemia, Brain; Hypoxia, Brain; Infarction, Middle Cerebral Artery; Male; Mice; Nerve Degeneration; Oxidative Stress; Time Factors

2009
Erythropoietin sustains cognitive function and brain volume after neonatal stroke.
    Developmental neuroscience, 2009, Volume: 31, Issue:5

    Neonatal stroke leads to mortality and severe morbidity, but there currently is no effective treatment. Erythropoietin (EPO) promotes cytoprotection and neurogenesis in the short term following brain injury; however, long-term cognitive outcomes and optimal dosing regimens have not been clarified. We performed middle cerebral artery occlusion in postnatal day 10 rats, which were treated with either a single dose of EPO (5 U/g, i.p.) immediately upon reperfusion, or 3 doses of EPO (1 U/g, i.p. each) at 0 h, 24 h, and 7 days after injury. At 3 months after injury, rats treated with 3 doses of EPO did not differ from shams in the Morris water maze, and generally performed better than either rats treated with a single dose or vehicle-treated injured rats. These multiple-dose-treated rats also had increases in hemispheric volume and its subregions. These results suggest that additional, later doses of EPO may be required for cell repair, proliferation, and long-term incorporation into neural networks after neonatal brain injury.

    Topics: Animals; Animals, Newborn; Brain; Cognition; Cytoprotection; Erythropoietin; Exploratory Behavior; Hypoxia-Ischemia, Brain; Infarction, Middle Cerebral Artery; Magnetic Resonance Imaging; Maze Learning; Memory; Neurons; Organ Size; Rats; Rats, Sprague-Dawley; Spatial Behavior

2009
Desferoxamine preconditioning protects against cerebral ischemia in rats by inducing expressions of hypoxia inducible factor 1 alpha and erythropoietin.
    Neuroscience bulletin, 2008, Volume: 24, Issue:2

    To investigate whether desferoxamine (DFO) preconditioning can induce tolerance against cerebral ischemia and its effect on the expression of hypoxia inducible factor 1alpha (HIF-1alpha) and erythropoietin (EPO) in vivo and in vitro.. Rat model of cerebral ischemia was established by middle cerebral artery occlusion with or without DFO administration. Infarct size was examined by TTC staining, and the neurological severity score was evaluated according to published method. Cortical neurons were cultured under ischemia stress which was mimicked by oxygen-glucose deprivation (OGD), and the neuron damage was assessed by MTT assay. Immunofluorescent staining was employed to detect the expressions of HIF-1alpha and EPO.. The protective effect induced by DFO (decreasing the infarction volume and ameliorating the neurological function) appeared at 2 d after administration of DFO (post-DFO), lasted until 7 d and disappeared at 14 d (P < 0.05); the most effective action was observed at 3 d post-DFO. DFO induced tolerance of cultured neurons against OGD: neuronal viability was increased 23%, 34%, 40%, 48% and 56% at 8 h, 12 h, 24 h, 36 h, and 48 h, respectively, post-DFO (P < 0.05). Immunofluorescent staining found that HIF-1alpha and EPO were upregulated in the neurons of rat brain at 3 d and 7 d post-DFO; increase of HIF-1alpha and EPO appeared in cultured cortex neurons at 36 h and 48 h post-DFO.. DFO induced tolerance against focal cerebral ischemia in rats, and exerted protective effect on OGD cultured cortical neurons. DFO significant induced the expression of HIF-1alpha and EPO both in vivo and in vitro. DFO preconditioning can protect against cerebral ischemia, which may be associated with the synthesis of HIF-1alpha and EPO.

    Topics: Animals; Brain Ischemia; Cells, Cultured; Cerebral Infarction; Deferoxamine; Disease Models, Animal; Erythropoietin; Fluorescent Antibody Technique; Hypoxia-Inducible Factor 1, alpha Subunit; Hypoxia-Ischemia, Brain; Infarction, Middle Cerebral Artery; Iron; Ischemic Preconditioning; Nerve Degeneration; Neurons; Rats; Rats, Sprague-Dawley; Siderophores; Time Factors; Treatment Outcome; Up-Regulation

2008
Effects of erythropoietin on blood-brain barrier disruption in focal cerebral ischemia.
    Pharmacology, 2008, Volume: 82, Issue:1

    This study was performed to test whether systemically administered erythropoietin (EPO) could attenuate the blood-brain barrier (BBB) disruption in focal ischemia. Rats were injected intraperitoneally with 2,500 IU/kg of recombinant human EPO or normal saline 24 h before middle cerebral artery (MCA) occlusion. The transfer coefficient (Ki) of 14C-alpha-aminoisobutyric acid was determined to measure the degree of BBB disruption 1 h after MCA occlusion. In the control animals, the Ki of the ischemic cortex (IC) was significantly higher than that of the contralateral cortex (CC; +128%, p = 0.0002). In the EPO-treated animals, the Ki of the IC was not significantly different from that of the CC and was significantly lower (-44%, p = 0.003) than that of the control animals. Our data suggest that MCA occlusion increased BBB disruption, and the disruption was attenuated with EPO pretreatment.

    Topics: Aminoisobutyric Acids; Animals; Blood-Brain Barrier; Capillary Permeability; Cerebral Cortex; Erythropoietin; Infarction, Middle Cerebral Artery; Injections, Intraperitoneal; Ischemic Attack, Transient; Male; Rats; Rats, Wistar; Recombinant Proteins

2008
Evaluation of erythropoietin effects on cerebral ischemia in rats.
    Neuro endocrinology letters, 2007, Volume: 28, Issue:2

    Majority of severe disabilities in adults are caused by stroke. The aim of our study is to learn the effects of erythropoietin (EPO), on infarct size in cerebral ischemia and to determine neurological behavioral scores and histopathological evaluation.. In this study 30 adult Sprague-Dawney rats were used. Cerebral ischemia was constituted by intraluminal filament method with a 4-0-nylon suture. Reperfusion was started after two hours of middle cerebral artery occlusion. The rats were randomly divided into two groups as follow: control and EPO groups. Saline 0.9% (0.5 ml/kg) and EPO (5 000 U/kg) was administered intraperitoneally in the groups. Three coronal slices in two millimeters thickness were obtained from cerebrum, cerebellum and brain stem, and were stained with a 2% solution of triphenyltetrazolium chloride. Transparent sheets were placed over each section and the areas of the brain and infarct were measured. The neurological scores were determined at 24th, 48th and 72nd hours after reperfusion.. Percent of ischemic area (%) in cerebrum, cerebellum and brain stem level in EPO groups were less than those of control group (p<0.0001). In addition, we determined that EPO group was better than controls of neurologic score and histopathologically after cerebral ischemia.. We concluded that EPO may decrease ischemic area in experimental cerebral ischemia in rats and it seems that EPO may be beneficial.

    Topics: Animals; Brain Ischemia; Drug Evaluation, Preclinical; Erythropoietin; Infarction, Middle Cerebral Artery; Injections, Intraperitoneal; Male; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Treatment Outcome

2007
Erythropoietin prevents blood brain barrier damage induced by focal cerebral ischemia in mice.
    Neurochemical research, 2007, Volume: 32, Issue:12

    Recombinant human erythropoietin (rhEPO), a neurovascular protective agent, therapeutically supports angiogenesis after stroke by enhancing endogenous up-regulation of vascular endothelial growth factor (VEGF). Increased VEGF expression has been characterized to negatively impact the integrity of the blood brain barrier (BBB), causing brain edema and secondary injury. The present study investigated the rhEPO-induced BBB protection after stroke and how it might be achieved by affecting VEGF pathway. rhEPO treatment (5,000 U/kg, i.p., 30 min before stroke and once a day for three days after stroke) reduced Evans blue leakage and brain edema after ischemia. The expression of the BBB integrity markers, occludin, alpha-catenin and beta-catenin, in the brain was preserved in animals received rhEPO. rhEPO up-regulated VEGF expression; however, the expression of VEGF receptor-2 (fetal liver kinase receptor, Flk-1) was significantly reduced in rhEPO-treated animals three days after stroke. We propose that, disregarding increased VEGF levels, rhEPO protects against ischemia-induced BBB damage at least partly by down-regulating Flk-1 expression and the response to VEGF signaling in the acute phase after stroke.

    Topics: Animals; Blood-Brain Barrier; Brain Edema; Cerebrovascular Circulation; Down-Regulation; Erythropoietin; Evans Blue; Fluorescent Antibody Technique; Infarction, Middle Cerebral Artery; Ischemic Attack, Transient; Male; Membrane Proteins; Mice; Microscopy, Confocal; Occludin; Recombinant Proteins; Stroke; Vascular Endothelial Growth Factor Receptor-2

2007
Post-ischemic treatment with erythropoietin or carbamylated erythropoietin reduces infarction and improves neurological outcome in a rat model of focal cerebral ischemia.
    British journal of pharmacology, 2007, Volume: 151, Issue:8

    Recombinant human erythropoietin (rhEPO; Epoetin-alpha; PROCRITtrade mark) has been shown to exert neuroprotective and restorative effects in a variety of CNS injury models. However, limited information is available regarding the dose levels required for these beneficial effects or the neuronal responses that may underlie them. Here we have investigated the dose-response to rhEPO and compared the effects of rhEPO with those of carbamylated rhEPO (CEPO) in a model of cerebral stroke in rats.. Rats subjected to embolic middle cerebral artery occlusion (MCAo) were treated with rhEPO or CEPO, starting at 6 h and repeated at 24 and 48 h, after MCAo. Cerebral infarct volumes were assessed at 28 days and neurological impairment at 7, 14, 21 and 28 days, post-MCAo.. rhEPO at dose levels of 500, 1150 or 5000 IU kg(-1) or CEPO at a dose level of 50 microg kg(-1) significantly reduced cortical infarct volume and reduced neurologic impairment. All doses of rhEPO, but not CEPO, produced a transient increase in haematocrit, while rhEPO and CEPO substantially reduced the number of apoptotic cells and activated microglia in the ischemic boundary region.. These data indicate that rhEPO and CEPO have anti-inflammatory and anti-apoptotic effects, even with administration at 6 h following embolic MCAo in rats. Taken together, these actions of rhEPO and CEPO are likely to contribute to their reduction of neurologic impairment following cerebral ischemia.

    Topics: Animals; Apoptosis; Blood-Brain Barrier; Brain Ischemia; Disease Models, Animal; Dose-Response Relationship, Drug; Enzyme-Linked Immunosorbent Assay; Epoetin Alfa; Erythropoietin; Hematinics; Hematocrit; Infarction, Middle Cerebral Artery; Male; Microglia; Neuroprotective Agents; Rats; Rats, Wistar; Recombinant Proteins; Stroke

2007
Neuroprotective effect of erythropoietin, and role of metallothionein-1 and -2, in permanent focal cerebral ischemia.
    Neuroscience, 2007, Aug-10, Volume: 148, Issue:1

    Metallothioneins (MTs) are small cysteine-rich proteins found widely throughout the mammalian body, including the CNS. MT-1 and -2 protect against reactive oxygen species and free radicals. We investigated the role of MT-1 and -2 using MT-1,-2 knockout (KO) mice. MT-1,-2 KO mice exhibited greater neuronal damage after permanent middle cerebral artery occlusion (MCAO) than wild-type mice. MT-2 mRNA was significantly increased at 6, 12, and 24 h after MCAO in the wild-type mouse brain [as detected by real-time reverse-transcription polymerase chain reaction (RT-PCR)], while MT-1 and MT-3 were decreased at 12 and 24 h. In an immunohistochemical study, MT expression displayed colocalization with glial fibrillary acidic protein (GFAP)-positive cells (astrocytes) in the penumbra area in wild-type mice. Since erythropoietin (EPO) has been reported to induce MT-1 and -2 gene expression in vitro, we examined its effect after permanent MCAO, and explored the possible underlying mechanism by examining MT-1 and -2 induction in vivo. In wild-type mice, EPO significantly reduced both infarct area and volume at 24 h after the ischemic insult. However, in MT-1,-2 KO mice EPO-treatment did not alter infarct volume (vs. vehicle-treatment). In wild-type mice at 6 h after EPO administration, real-time RT-PCR revealed increased MT-1 and -2 mRNA expression in the cerebral cortex (without MCAO). Further, MT-1 and -2 immunoreactivity was increased in the cortex of EPO-treated mice. These findings indicate that MTs are induced, and may be neuroprotective against neuronal damage, after MCAO. Furthermore, EPO is neuroprotective in vivo during permanent MCAO, and this may be at least partly mediated by MTs.

    Topics: Animals; Astrocytes; Brain; Brain Infarction; Brain Ischemia; Cytoprotection; Erythropoietin; Gene Expression Regulation; Glial Fibrillary Acidic Protein; Infarction, Middle Cerebral Artery; Male; Metallothionein; Metallothionein 3; Mice; Mice, Inbred C57BL; Mice, Knockout; Nerve Degeneration; Neuroprotective Agents; RNA, Messenger; Up-Regulation

2007
Erythropoietin enhances long-term neuroprotection and neurogenesis in neonatal stroke.
    Developmental neuroscience, 2007, Volume: 29, Issue:4-5

    Neonatal stroke leads to mortality and severe morbidity, but there is no effective treatment currently available. Erythropoietin (EPO) has been shown to promote cytoprotection and neurogenesis and decrease subventricular zone morphologic changes following brain injury. The long-term cellular response to EPO has not been defined, and local changes in cell fate decision may play a role in functional improvement. We performed middle cerebral artery occlusion in P10 rats. EPO treatment (5 U/g i.p.) significantly preserved hemispheric brain volume 6 weeks after injury. Furthermore, EPO increased the percentage of newly generated neurons while decreasing newly generated astrocytes following brain injury, without demonstrating long-term differences in the subventricular zone. These results suggest that EPO may neuroprotect and direct cell fate toward neurogenesis and away from gliogenesis in neonatal stroke.

    Topics: Animals; Animals, Newborn; Astrocytes; Cell Differentiation; Cell Proliferation; Erythropoietin; Gliosis; Hypoxia, Brain; Infarction, Middle Cerebral Artery; Nerve Regeneration; Neurons; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recovery of Function; Stem Cells; Stroke; Time; Treatment Outcome

2007
A critical role of erythropoietin receptor in neurogenesis and post-stroke recovery.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2006, Jan-25, Volume: 26, Issue:4

    Erythropoietin (EPO) is the principal growth factor regulating the production of red blood cells. Recent studies demonstrated that exogenous EPO acts as a neuroprotectant and regulates neurogenesis. Using a genetic approach, we evaluate the roles of endogenous EPO and its classical receptor (EPOR) in mammalian neurogenesis. We demonstrate severe and identical embryonic neurogenesis defects in animals null for either the Epo or EpoR gene, suggesting that the classical EPOR is essential for EPO action during embryonic neurogenesis. Furthermore, by generating conditional EpoR knock-down animals, we demonstrate that brain-specific deletion of EpoR leads to significantly reduced cell proliferation in the subventricular zone and impaired post-stroke neurogenesis. EpoR conditional knockdown leads to a specific deficit in post-stroke neurogenesis through impaired migration of neuroblasts to the peri-infarct cortex. Our results suggest that both EPO and EPOR are essential for early embryonic neural development and that the classical EPOR is important for adult neurogenesis and for migration of regenerating neurons during post-injury recovery.

    Topics: Animals; Brain; Cell Division; Cell Lineage; DNA Replication; Erythropoietin; Gene Expression Regulation, Developmental; In Situ Hybridization; Infarction, Middle Cerebral Artery; Integrases; Mesoderm; Mice; Mice, Inbred C57BL; Mice, Knockout; Nerve Tissue Proteins; Neural Crest; Neural Tube Defects; Neuroepithelial Cells; Neurons; Receptors, Erythropoietin; Regeneration; Stem Cells; Viral Proteins

2006
Expression of the gene encoding the pro-apoptotic BNIP3 protein and stimulation of hypoxia-inducible factor-1alpha (HIF-1alpha) protein following focal cerebral ischemia in rats.
    Neurochemistry international, 2006, Volume: 48, Issue:8

    Hypoxia is a common cause of cell death and is implicated in many disease processes including stroke and chronic degenerative disorders. In response to hypoxia, cells express a variety of genes which allow adaptation to altered metabolic demands, decreased oxygen demands, and the removal of irreversibly damaged cells. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that regulates the adaptive response to hypoxia in cells. In this study, we reported an early, time-related, gradual up-regulation of HIF-1alpha, and a moderate increase in vascular endothelial growth factor (VEGF)- and erythropoietin (Epo)-levels following transient focal ischemia. Moreover, we demonstrated, for the first time a specific localization of the pro-apoptotic regulator BNIP3 in striatal and cortical neurons after transient focal ischemia in rats. Prolonged intranuclear BNIP3 immunoreactivity was associated with delayed neuronal death. Experiments showed protein increases on Western blots of brain tissue with peaks at 48h after ischemia. Epo responds to ischemia in an early stage, whereas VEGF and BNIP3 accumulate in cells at later times after ischemia. This suggests the possibility that BH3-only proteins might be one of the major downstream effectors of HIF-1alpha in hypoxic cell death. These findings open the possibility that the hypoxia-regulated pro-apoptotic protein BNIP3 enters the nucleus and could interact with other proteins involved in DNA structure, transcription or mRNA splicing after focal brain ischemia.

    Topics: Active Transport, Cell Nucleus; Animals; Apoptosis; Brain Ischemia; Cell Nucleus; Cerebral Cortex; Cerebral Infarction; Corpus Striatum; Disease Models, Animal; Erythropoietin; Gene Expression Regulation; Hypoxia-Inducible Factor 1, alpha Subunit; Infarction, Middle Cerebral Artery; Male; Membrane Proteins; Mitochondrial Proteins; Nerve Degeneration; Neurons; Proto-Oncogene Proteins; Rats; Rats, Wistar; Signal Transduction; Time Factors; Up-Regulation; Vascular Endothelial Growth Factor A

2006
Cerebral ischemic tolerance induced by 3-nitropropionic acid is associated with increased expression of erythropoietin in rats.
    Journal of Huazhong University of Science and Technology. Medical sciences = Hua zhong ke ji da xue xue bao. Yi xue Ying De wen ban = Huazhong keji daxue xuebao. Yixue Yingdewen ban, 2006, Volume: 26, Issue:4

    To examine the changes in erythropoietin (Epo) protein and its mRNA expression in rat brain subjected to focal ischemia and possible mechanism of the preconditioning of mitochondrial toxin 3-nitropropionic acid (3-NPA), rats were administrated either vehicle or 3-NPA at a dose of 20 mg/kg, intraperitoneally (ip), 3 days prior to a 2-h middle cerebral artery occlusion followed by 24-h reperfusion. Infarct volumes were measured by using 2, 3, 5 triphenylte trazolinm chloride (TTC) staining, and Epo protein and its mRNA levels were assessed by immunohistochemistry and reverse transcriptase polymerase chain reaction (RT-PCR), respectively. Our results showed that after reperfusion, Epo was found to be expressed extensively in the rat brain. It was most apparent in the basal nuclei and hippocampus, and was, to some extent, present in cortex. Preconditioning with 3-NPA caused a reduction in infarct volume. The expression of both Epo protein and mRNA increased significantly in the different brain areas in the 3-NPA pretreated group as compared with the non-pretreated ischemia model group. These results suggested that preconditioning with low dose 3-NPA could induce ischemic tolerance and neuro-protective effects by increasing the Epo expression in the ischemic and ischemia-related areas.

    Topics: Animals; Erythropoietin; Immunohistochemistry; Infarction, Middle Cerebral Artery; Ischemic Preconditioning; Male; Nitro Compounds; Propionates; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger

2006
Erythropoietin after focal cerebral ischemia activates the Janus kinase-signal transducer and activator of transcription signaling pathway and improves brain injury in postnatal day 7 rats.
    Pediatric research, 2005, Volume: 57, Issue:4

    Erythropoietin (Epo) plays a central role in erythropoiesis but also has neuroprotective properties. Recently, Epo-related neuroprotective studies used a hypoxic-ischemic neonatal model, which is different from focal stroke, a frequent cause of neonatal brain injury. We report on the effects of Epo treatment given after focal stroke and its potential neuroprotective mechanisms in postnatal day 7 rats with focal cerebral ischemia (FCI) achieved by occlusion of the middle cerebral artery. The experimental groups included sham operation, FCI plus vehicle, and FCI plus Epo. In the Epo-treated group, pups received a single intraperitoneal injection of 1000 U/kg 15 min after FCI or three injections of 100, 1000, or 5000 U/kg, starting at 15 min and repeated at 1 and 2 d after FCI. Epo treatment produced significant reductions in the mean infarct area and volume at 1 and 3 d after FCI, demonstrated by 2,3,5-triphenyltetrazolium chloride staining. Terminal deoxynucleotidyltransferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL) staining showed a markedly reduced number of TUNEL-positive cells in the Epo-treated group when compared with the vehicle control 3 d after FCI (p<0.01). The most effective dose after FCI was 1000 U/kg for 3 d. Immunoanalyses showed that Epo induced a significant increase in phosphorylated Janus kinase 2 and signal transducer and activator of transcription-5 expressions at 1 and 3 d and up-regulated Bcl-xL expression by 24 h after FCI but did not affect Epo receptor or NF-kappaB expression. In conclusion, Epo given after FCI in neonatal rats provides significant neuroprotection, mediated possibly by activation of the Janus kinase-signal transducer and activator of transcription-Bcl-xL signaling pathways.

    Topics: Animals; Animals, Newborn; bcl-X Protein; Brain Ischemia; Cerebral Cortex; DNA-Binding Proteins; Erythropoietin; Female; In Situ Nick-End Labeling; Infarction, Middle Cerebral Artery; Janus Kinase 2; Milk Proteins; NF-kappa B; Pregnancy; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-bcl-2; Rats; Rats, Sprague-Dawley; Receptors, Erythropoietin; Signal Transduction; STAT5 Transcription Factor; Trans-Activators

2005
Neuroprotective effect of darbepoetin alfa, a novel recombinant erythropoietic protein, in focal cerebral ischemia in rats.
    Stroke, 2005, Volume: 36, Issue:5

    Darbepoetin alfa is a novel erythropoiesis-stimulating protein developed for treating anemia. In animal models, exogenous recombinant human erythropoietin has been reported to be beneficial in treating experimental cerebral ischemia. In this study, we determined whether darbepoetin alfa would protect in a rat model of transient focal cerebral ischemia.. Rats received 2-hour middle cerebral artery suture-occlusion. The drug (darbepoetin alfa, 10 microg/kg) or vehicle was administered intraperitoneally 2 hours after onset of middle cerebral artery occlusion. Animals were allowed to survive for 3 or 14 days. Behavioral tests were performed sequentially. Infarct volumes and brain swelling were determined.. Darbepoetin alfa-treated rats showed improved neuroscores relative to vehicle-treated animals beginning within 1 hour of treatment and persisting throughout the 14-day survival period. Darbepoetin alfa significantly reduced corrected total (cortical + subcortical) infarct volume (56.3+/-20.6 and 110.8+/-6.8 mm3, respectively) and total infarct areas at multiple levels compared with vehicle in the 14-day survival group. Brain swelling was not affected by treatment.. Darbepoetin alfa confers behavioral and histological neuroprotection after focal ischemia in rats.

    Topics: Animals; Behavior, Animal; Brain Edema; Brain Ischemia; Darbepoetin alfa; Erythropoietin; Infarction, Middle Cerebral Artery; Male; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recombinant Proteins

2005
Hypoxia-inducible factor 1alpha and erythropoietin upregulation with deferoxamine salvage after neonatal stroke.
    Experimental neurology, 2005, Volume: 195, Issue:2

    Treatment with deferoxamine (DFO) is protective against focal ischemia with global hypoxia when given as a preconditioning stimulus in neonatal rodents. DFO acts as an iron chelator and may stabilize HIF1alpha. Therefore, we hypothesized that DFO would protect against pure ischemia-reperfusion injury when given after the insult and that the protection would be associated with expression of hypoxia-inducible factor 1alpha (HIF1alpha) and downstream target genes such as erythropoietin (Epo). To test these hypotheses, we performed middle cerebral artery (MCA) occlusion in postnatal day 10 (P10) rats for 1.5 h followed by treatment with DFO or vehicle upon reperfusion. Preserved brain volumes were measured with cresyl violet staining 1 week after the insult. HIF1alpha and Epo expression were determined by Western blot and immunocytochemical analyses at different time points after injury. We found that DFO treatment preserved brain volumes when compared to vehicle (P < 0.05). In DFO-treated ischemic cortices, HIF1alpha expression peaked early, while Epo expression was seen in two phases and in different cell populations. Epo immunoreactivity colocalized with neuronal markers at 8 h but with astrocytic markers at 1 week. These results suggest that DFO is protective when administered after neonatal ischemic stroke and that this protection may be like that afforded by preconditioning through the upregulation of similar downstream pathways.

    Topics: Animals; Animals, Newborn; Blotting, Western; Deferoxamine; Disease Models, Animal; Erythropoietin; Fluorescent Antibody Technique; Glial Fibrillary Acidic Protein; Immunohistochemistry; Infarction, Middle Cerebral Artery; Iron Chelating Agents; Phosphopyruvate Hydratase; Rats; Time Factors; Up-Regulation

2005
Intranasal recombinant human erythropoietin protects rats against focal cerebral ischemia.
    Neuroscience letters, 2005, Oct-14, Volume: 387, Issue:1

    Erythropoietin (EPO) is a hematopoietic growth factor with tissue-protective properties, and can protect animals from cerebral ischemic injury. However, the central nervous effects of EPO as a glycoprotein is limited by the potential complication resulted from its erythropoietic activity and the problem of the penetration through blood-brain barrier (BBB). To avoid these limitations, in this study we administered recombinant human EPO (rhEPO) intranasally (i.n.) to evaluate its neuroprotective effect in the rats with focal cerebral ischemia induced by middle cerebral artery occlusion (MCAO). We found that rhEPO i.n. at doses of 4.8, 12 and 24 U (administered 10 min after MCAO and 1h after reperfusion) reduced infarct volume, brain swelling and cell damage in the ischemic hemispheres, and improved behavioral dysfunction 24 h after cerebral ischemia. Intraperitoneal rhEPO (5000 U/kg) also showed the protective effect, but the heat-inactivated rhEPO did not show any effect. Thus, intranasal administration of relatively small doses of rhEPO protects rats from acute injury after focal cerebral ischemia, suggesting that intranasal rhEPO may be a more effective and safer administration route for treatments of ischemic or other brain diseases.

    Topics: Administration, Intranasal; Animals; Brain Edema; Brain Ischemia; Cerebral Infarction; Disease Models, Animal; Dose-Response Relationship, Drug; Erythropoietin; Humans; Infarction, Middle Cerebral Artery; Male; Nerve Degeneration; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Recovery of Function; Treatment Outcome

2005
Treatment of stroke with erythropoietin enhances neurogenesis and angiogenesis and improves neurological function in rats.
    Stroke, 2004, Volume: 35, Issue:7

    Erythropoietin (EPO) promotes proliferation and differentiation of erythroid progenitors and the survival of maturing erythroid cells. Here, we investigated the role of EPO in brain repair after stroke.. Rats were treated with recombinant human EPO (rhEPO) at 24 hours after the onset of embolic stroke. An array of behavior tests was performed. Rats were euthanized 28 days after stroke for measurements of infarct volume, angiogenesis, and neurogenesis. In vitro, neurospheres derived from the subventricular zone (SVZ) of the rat and cerebral endothelial cells derived from the mouse were treated with rhEPO. Capillary-like tube formation and neuronal differentiation were measured.. Treatment with rhEPO significantly improved functional recovery, along with increases in density of cerebral microvessels at the stroke boundary and numbers of BrdU, doublecortin, and nestin immunoreactive cells in the SVZ. rhEPO treatment significantly increased brain levels of vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF). In vitro, rhEPO enhanced capillary tube formation of cerebral endothelial cells, which was inhibited by a specific VEGF receptor 2 antagonist (SU1498). Incubation of neurospheres derived from stroke SVZ with anti-EPO neutralizing antibody inhibited neurogenesis, whereas incubation of stroke-derived neurospheres with rhEPO enhanced neurogenesis.. Our data suggest that EPO-increased VEGF and BDNF may be involved in angiogenesis and neurogenesis, which could contribute to functional recovery.

    Topics: Angiogenesis Inducing Agents; Animals; Brain; Brain-Derived Neurotrophic Factor; Doublecortin Protein; Erythropoietin; Infarction, Middle Cerebral Artery; Male; Microcirculation; Models, Animal; Neovascularization, Physiologic; Nerve Regeneration; Neuronal Plasticity; Rats; Rats, Wistar; Recombinant Proteins; Stroke; Vascular Endothelial Growth Factor A

2004
Transient middle cerebral artery occlusion influence on systemic oxygen homeostasis and erythropoiesis in Wistar rats.
    Stroke, 2004, Volume: 35, Issue:8

    Systemic hypoxia is a common complication in stroke patients and may exacerbate ischemic brain damage. Expression of the hypoxia-inducible cytokine erythropoietin (Epo) is upregulated in the brain in both stroke patients and in animal stroke models and exerts local neuroprotective effects in the ischemic brain. Epo is also well known to stimulate red blood cell (RBC) production. The purpose of the present study was to evaluate whether poststroke systemic hypoxia is present in the rat model and whether it is associated with increased peripheral Epo and RBC production.. Wistar rats underwent 1-hour transient middle cerebral artery occlusion (MCAO) under mechanical ventilation, followed by reperfusion without further ventilation. Groups of MCAO and sham-operated animals were evaluated at extended times after reperfusion for assessment of arterial blood gases, plasma Epo, and complete blood count.. Arterial oxygen saturation was significantly lower in the infarct group between 6 and 24 hours after reperfusion (P=0.0005), and plasma Epo levels were increased 6 hours after reperfusion (P<0.05). RBC counts and hematocrit were transiently increased 2 to 7 days after reperfusion in animals with MCAO compared with sham. Maximal increases were seen at day 7 (22% and 16% increases of RBC count and hematocrit, respectively; P<0.001). In contrast, the white blood cell counts in animals with MCAO decreased by >30% in the same time period.. Plasma Epo levels, RBC counts, and hematocrit are all increased in response to systemic hypoxia after cerebral ischemia in rats.

    Topics: Animals; Disease Models, Animal; Erythrocyte Count; Erythropoiesis; Erythropoietin; Hematocrit; Homeostasis; Hypoxia; Infarction, Middle Cerebral Artery; Male; Oxygen; Rats; Rats, Wistar; Stroke

2004
Hypoxia-induced stroke tolerance in the mouse is mediated by erythropoietin.
    Stroke, 2003, Volume: 34, Issue:8

    Cellular response to hypoxia is mainly controlled by hypoxia-inducible factor 1 (HIF-1). The HIF-1 target gene erythropoietin (EPO) has been described as neuroprotective. Thus, we hypothesize EPO to be an essential mediator of protection in hypoxic preconditioning.. We randomized Sv129 mice into groups for different pretreatments, different hypoxia-ischemia intervals, or different durations of ischemia. For hypoxic preconditioning, the animals were exposed to a hypoxic gas mixture (8% O2 and 92% N2) for 30, 60, 180, 300, or 360 minutes. At 0, 24, 48, 72, or 144 hours later, we performed middle cerebral artery occlusion and allowed reperfusion after 30, 45, 60, or 120 minutes, or occlusion was left to be permanent. We studied EPO gene expression in brain tissue with a real-time reverse transcriptase-polymerase chain reaction and measured HIF-1 DNA-binding activity with an electrophoretic mobility shift assay. To block endogenously produced EPO, we instilled soluble EPO receptor into the cerebral ventricle.. Hypoxic preconditioning for 180 or 300 minutes induced relative tolerance to transient focal cerebral ischemia, as evidenced by a reduction of infarct volumes to 75% or 54% of the control, respectively. Hypoxic pretreatment was effective only when applied 48 or 72 hours before middle cerebral artery occlusion. Sixty minutes after hypoxia, we found a marked activation of HIF-1 DNA-binding activity and a 7-fold induction of EPO transcription. Infusion of soluble EPO receptor significantly reduced the protective effect of hypoxic pretreatment by 40%.. Endogenously produced EPO is an essential mediator of ischemic preconditioning.

    Topics: Animals; Brain; Cerebral Infarction; Disease Models, Animal; DNA; DNA-Binding Proteins; Electrophoretic Mobility Shift Assay; Erythropoietin; Female; Hippocampus; Hypoxia; Hypoxia-Inducible Factor 1; Hypoxia-Inducible Factor 1, alpha Subunit; Infarction, Middle Cerebral Artery; Injections, Intraventricular; Ischemic Preconditioning; Male; Mice; Mice, Inbred Strains; Nuclear Proteins; Receptors, Erythropoietin; Reperfusion; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Stroke; Time Factors; Transcription Factors

2003
Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis.
    The Journal of experimental medicine, 2003, Sep-15, Volume: 198, Issue:6

    Ischemic brain injury resulting from stroke arises from primary neuronal losses and by inflammatory responses. Previous studies suggest that erythropoietin (EPO) attenuates both processes. Although EPO is clearly antiapoptotic for neurons after experimental stroke, it is unknown whether EPO also directly modulates EPO receptor (EPO-R)-expressing glia, microglia, and other inflammatory cells. In these experiments, we show that recombinant human EPO (rhEPO; 5,000 U/kg body weight, i.p.) markedly reduces astrocyte activation and the recruitment of leukocytes and microglia into an infarction produced by middle cerebral artery occlusion in rats. In addition, ischemia-induced production of the proinflammatory cytokines tumor necrosis factor, interleukin 6, and monocyte chemoattractant protein 1 concentration is reduced by >50% after rhEPO administration. Similar results were also observed in mixed neuronal-glial cocultures exposed to the neuronal-selective toxin trimethyl tin. In contrast, rhEPO did not inhibit cytokine production by astrocyte cultures exposed to neuronal homogenates or modulate the response of human peripheral blood mononuclear cells, rat glial cells, or the brain to lipopolysaccharide. These findings suggest that rhEPO attenuates ischemia-induced inflammation by reducing neuronal death rather than by direct effects upon EPO-R-expressing inflammatory cells.

    Topics: Animals; Apoptosis; Brain Ischemia; Cells, Cultured; Coculture Techniques; Cytokines; Erythropoietin; Humans; Infarction, Middle Cerebral Artery; Inflammation; Lipopolysaccharides; Male; Neuroglia; Neurons; Neuroprotective Agents; Rats; Receptors, Erythropoietin; Recombinant Proteins; Tumor Necrosis Factor-alpha

2003