Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Environ Sci Pollut Res Int ; 27(22): 27926-27936, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32405940

ABSTRACT

Recycled moisture, mainly originated from evapotranspiration (surface evaporation and transpiration), is the main sources of precipitation. Influenced on the different regional/local environments, the contributions of recycled moisture to precipitation present as different proportions. Recycled moisture has an important impact on the hydrological cycle, further occurred a series of environmental effect for regional/local. Aimed to estimate the contribution of recycled moisture to precipitation in an enclosed basin, Guanzhong Basin of northern China, precipitation and lake/reservoir samples were collected. The isotope ratio analysis was done for the summer season, and a three-component mixing model based on the stable hydrogen and oxygen isotopes was applied. The results indicated that the averaged contribution of recycled moisture to precipitation was 17.44% in Guanzhong Basin of northern China, while the mean proportions of surface evaporation moisture and transpiration moisture were found to be 0.38% and 16.97%, respectively. Comparatively, most of the recycled moisture mainly comes from transpiration moisture rather than evaporation moisture, suggesting that transpiration moisture from cropland, vegetation, and plants instead of evaporation is dominant in moisture recycling of the Guanzhong Basin.


Subject(s)
Environmental Monitoring , Rain , China , Oxygen Isotopes/analysis , Seasons
2.
J Neurointerv Surg ; 9(12): 1248-1252, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27899518

ABSTRACT

OBJECTIVE: To examine the hypothesis that IA reperfusion with iso-osmolar iodixanol, low-osmolar iopamidol, or saline causes different effects on MR signal changes and pathologic cut-brain section related to hemorrhagic transformation (HT) or iodinated radiographic contrast media (IRCM) deposition. METHODS: Infarct was induced in 30 rats by middle cerebral artery suture occlusion. Reperfusion was performed after 5 hours with iso-osmolar iodixanol (n=9), low-osmolar iopamidol (n=12) or saline (n=9). MR images were obtained immediately after reperfusion and rats were sacrificed at 24 hours. Hypointense areas within the infarction on T2-weighted (T2-WI) or gradient echo (GRE) images were recorded and compared with HT on pathology. Fisher's exact test was used for proportions, and receiver operator curve analysis to evaluate MRI discrimination of hemorrhage. RESULTS: Two types of HT were noted on pathology: confluent >0.2 mm petechial hemorrhage (PeH, 78%) or well-defined ≤0.2 mm hemorrhagic focus (HF, 22%). PeH was least common in the iodixanol subgroup (p<0.02). HF was more common in the IRCM group. Hypointense areas on T2-WI but not on GRE were significantly more common in the IRCM group (p<0.05). Hypointense areas on T2-WI and GRE discriminated HT (area under the curve: 0.714, p<0.002). CONCLUSIONS: IRCM and saline induced different MRI signal and pathologic patterns in our sample. We postulate that T2 hypointensity with no GRE hypointensity might be associated with IRCM deposition; and decreased frequency of PeH after iodixanol infusion and the presence of HF almost exclusively in the IRCM group might represent a direct/indirect effect of contrast infusion/deposition in the brain parenchyma after reperfusion. Our results support previous observations in IMS III and are hypothesis generating.


Subject(s)
Cerebral Hemorrhage/diagnostic imaging , Contrast Media/administration & dosage , Disease Models, Animal , Infarction, Middle Cerebral Artery/diagnostic imaging , Magnetic Resonance Imaging/trends , Triiodobenzoic Acids/administration & dosage , Animals , Brain/diagnostic imaging , Brain/drug effects , Cerebral Hemorrhage/complications , Contrast Media/adverse effects , Infarction, Middle Cerebral Artery/etiology , Infusions, Intraventricular , Magnetic Resonance Imaging/methods , Male , Rats , Rats, Sprague-Dawley , Reperfusion , Triiodobenzoic Acids/adverse effects
3.
J Cereb Blood Flow Metab ; 34(2): 185-99, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24281743

ABSTRACT

Hemorrhagic transformation (HT) is a common complication of ischemic stroke that is exacerbated by thrombolytic therapy. Methods to better prevent, predict, and treat HT are needed. In this review, we summarize studies of HT in both animals and humans. We propose that early HT (<18 to 24 hours after stroke onset) relates to leukocyte-derived matrix metalloproteinase-9 (MMP-9) and brain-derived MMP-2 that damage the neurovascular unit and promote blood-brain barrier (BBB) disruption. This contrasts to delayed HT (>18 to 24 hours after stroke) that relates to ischemia activation of brain proteases (MMP-2, MMP-3, MMP-9, and endogenous tissue plasminogen activator), neuroinflammation, and factors that promote vascular remodeling (vascular endothelial growth factor and high-moblity-group-box-1). Processes that mediate BBB repair and reduce HT risk are discussed, including transforming growth factor beta signaling in monocytes, Src kinase signaling, MMP inhibitors, and inhibitors of reactive oxygen species. Finally, clinical features associated with HT in patients with stroke are reviewed, including approaches to predict HT by clinical factors, brain imaging, and blood biomarkers. Though remarkable advances in our understanding of HT have been made, additional efforts are needed to translate these discoveries to the clinic and reduce the impact of HT on patients with ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Cerebral Hemorrhage/metabolism , Stroke/metabolism , Animals , Biomarkers/metabolism , Blood-Brain Barrier/pathology , Brain Ischemia/complications , Brain Ischemia/pathology , Brain Ischemia/therapy , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/prevention & control , Enzyme Activation/drug effects , HMGB1 Protein/biosynthesis , Humans , Peptide Hydrolases/metabolism , Protease Inhibitors/therapeutic use , Risk Factors , Signal Transduction/drug effects , Stroke/complications , Stroke/pathology , Stroke/therapy , Thrombolytic Therapy/methods , Time Factors , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , src-Family Kinases/metabolism
4.
BMC Genomics ; 12: 499, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21988864

ABSTRACT

BACKGROUND: Since normal brain function depends upon continuous oxygen delivery and short periods of hypoxia can precondition the brain against subsequent ischemia, this study examined the effects of brief hypoxia on the whole genome transcriptional response in adult mouse brain. RESULT: Pronounced changes of gene expression occurred after 3 hours of hypoxia (8% O(2)) and after 1 hour of re-oxygenation in all brain regions. The hypoxia-responsive genes were predominantly up-regulated in hindbrain and predominantly down-regulated in forebrain - possibly to support hindbrain survival functions at the expense of forebrain cognitive functions. The up-regulated genes had a significant role in cell survival and involved both shared and unshared signaling pathways among different brain regions. Up-regulation of transcriptional signaling including hypoxia inducible factor, insulin growth factor (IGF), the vitamin D3 receptor/retinoid X nuclear receptor, and glucocorticoid signaling was common to many brain regions. However, many of the hypoxia-regulated target genes were specific for one or a few brain regions. Cerebellum, for example, had 1241 transcripts regulated by hypoxia only in cerebellum but not in hippocampus; and, 642 (54%) had at least one hepatic nuclear receptor 4A (HNF4A) binding site and 381 had at least two HNF4A binding sites in their promoters. The data point to HNF4A as a major hypoxia-responsive transcription factor in cerebellum in addition to its known role in regulating erythropoietin transcription. The genes unique to hindbrain may play critical roles in survival during hypoxia. CONCLUSION: Differences of forebrain and hindbrain hypoxia-responsive genes may relate to suppression of forebrain cognitive functions and activation of hindbrain survival functions, which may coordinately mediate the neuroprotection afforded by hypoxia preconditioning.


Subject(s)
Brain/metabolism , Hypoxia , Transcriptome , Animals , Binding Sites , Cerebellum/metabolism , Genome , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Time Factors , Up-Regulation
5.
Neurol Res ; 33(4): 433-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21535944

ABSTRACT

OBJECTIVE: To investigate recombinant human TIMP-1 ((125)I-rhTIMP-1) half-life in blood and its distribution in rat brain tissue after cerebral ischemia/reperfusion as part of a therapeutic development paradigm. METHOD: A suture model of the middle cerebral artery occlusion was used. (125)I-labeled rhTIMP-1 at 60 µg/kg (11.23 µCi/µg) was administered to rats intravenously at the beginning of reperfusion. Blood and brain tissue were collected. The radioactivity was detected with a gamma counter and analyzed by autoradiography. RESULTS: The blood half-life T(1/2) of (125)I-rhTIMP-1 was 42.2 hours. Thirty minutes after (125)I-rhTIMP-1 administration, an increased accumulation of (125)I-rhTIMP-1 in the ischemic hemisphere was observed. The maximum brain tissue concentration C(max) was 26.1 ng/g at 1.5 hours in the striatum and 13.9 ng/g at 5 hours in the cortex when the uptake percentage of brain tissue to blood was 6.1±0.4 and 6.7±2.1%, respectively. The cortex and striatum elimination half-lives T(1/2) were 45.3 and 39.2 hours, respectively. Electrophoretic analysis of ischemic samples for (125)I-rhTIMP-1 showed a clear 28 kDa band 1.5 hours after (125)I-rhTIMP-1 administration in the cortex and striatum. The intensity of the 28 kDa band decreased after 3.0 hours of the administration. Some (125)I-rhTIMP-1 maintained its molecular integrity for 8.5 hours in ischemic striatum after reperfusion. DISCUSSION: (125)I-labeled rhTIMP-1 was distributed quickly into ischemic brain tissue and had a slow elimination in both blood and brain tissue. These results, along with other studies suggesting therapeutic benefits, will aid in the development of TIMP-1 for protecting ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Protease Inhibitors/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Reperfusion Injury/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacokinetics , Animals , Brain/blood supply , Brain/drug effects , Brain Ischemia/drug therapy , Disease Models, Animal , Half-Life , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Iodine Radioisotopes , Male , Metabolic Clearance Rate , Protease Inhibitors/blood , Protease Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/therapeutic use , Reperfusion Injury/drug therapy , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/therapeutic use
6.
Acta Neurochir Suppl ; 110(Pt 1): 157-62, 2011.
Article in English | MEDLINE | ID: mdl-21116932

ABSTRACT

INTRODUCTION: We have previously shown that novel oxidation products of Bilirubin, called Bilirubin oxidation products (BOXes), are found in humans and animal models post subarachnoid hemorrhage. We have also proposed that BOXes may play a role in the pathogenesis and clinical complications post SAH. In this study we report on the direct toxicity effects of BOXes on rat brain. METHODS: Identical volumes of either vehicle (normal saline) or BOXes (30 µl of a 20 µM solution) were applied above the dura through a cranial window of young (approximately 7-13 weeks) and aged (approximately 12-18 months) adult male Sprague Dawley rats (Charles River, Wilmington, MA, USA). To determine the extent of BOX-mediated injury, histology and immunocytochemistry were performed at 1, 2, 4, and 7 days post-surgical application of BOXes. We assessed the area of stress gene induction of HSP25/27 and HSP32. Immunohistochemistry was performed using standard avidin-biotin techniques. A monoclonal antibody to HSP25/27 (StressGen, Victoria, British Columbia, Canada), a monoclonal antibody to HSP32/HO-1 (StressGen), and a polyclonal HSP 32/HO-1 antibody were used for the immunocytochemistry. RESULTS: A single dose of BOXes produced substantial increases in HSP25 and HO-1 in the aged rats at all early time points (≤4 days). After 7 days all groups were not significantly different than saline control. Young rats were resistant to BOXes effects compared to saline control with trends towards increased stress gene expression caused by BOXes that did not reach statistical significance. CONCLUSION: We conclude from these studies that BOXes have direct effects on stress gene expression of the cortex post single dose application and that this can be seen for several days with apparent resolution at about 7 days. If BOXes are produced at similar levels in patients, the latency and duration of some SAH complications are consistent with these results.


Subject(s)
Aging , Antioxidants/metabolism , Bilirubin/metabolism , Brain/metabolism , Subarachnoid Hemorrhage/pathology , Analysis of Variance , Animals , Antioxidants/chemistry , Bilirubin/chemistry , Brain/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , HSP27 Heat-Shock Proteins/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Male , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Spectrophotometry/methods , Time Factors
7.
J Neurochem ; 115(5): 1266-76, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20883517

ABSTRACT

Matrix metalloproteinases (MMPs) play an important role in reperfusion-induced brain injury following ischemia. To define the effects of peroxynitrite decomposition catalyst on MMP activation and neurovascular reperfusion injury, 5,10,15,20-tetrakis (2,4,6-trimethyl-3,5-disulfonatophenyl)-porphyrin iron (III) (FeTMPyP) was administered intravenously 30 min prior to reperfusion following a middle cerebral artery occlusion. Activation of MMP was assessed by in situ and gel zymography. Neurovascular injury was assessed using endothelial barrier antigen, collagen IV immunohistochemistry and Cresyl violet staining. Results were compared with sham and ischemia alone groups. We found that administration of FeTMPyP just before reperfusion after ischemia inhibited MMP-9 activation and total MMP-2 increases in the cortex and decreased active MMP-9 along with the total amounts of active MMP-9 and active MMP-2 in the striatum. Reperfusion-induced injury to the basal lamina of collagen IV-immunopositive microvasculature and neural cells in cortex and striatum was ameliorated by FeTMPyP. Losses of blood vessel endothelium produced by ischemia or reperfusion were also decreased in the cortex. These results suggest that administration of FeTMPy prior to reperfusion decreases MMP activation and neurovascular injury after prolonged cerebral ischemia. This strategy may be useful for future therapies targeted at preventing breakdown of the blood-brain barrier and hemorrhagic transformation.


Subject(s)
Cerebral Infarction/prevention & control , Ferric Compounds/therapeutic use , Hematinics/therapeutic use , Matrix Metalloproteinases/metabolism , Metalloporphyrins/therapeutic use , Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Astrocytes/drug effects , Astrocytes/pathology , Basement Membrane/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Corpus Striatum/pathology , Disease Models, Animal , Enzyme Activation/drug effects , Ferric Compounds/pharmacology , Hematinics/pharmacology , Infarction, Middle Cerebral Artery/complications , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Metalloporphyrins/pharmacology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Reperfusion Injury/etiology , Time Factors
8.
Stroke ; 41(5): 1013-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20360541

ABSTRACT

BACKGROUND AND PURPOSE: Observations in human interventional stroke treatment led us to hypothesize that iodinated radiographic contrast material use may contribute to intracerebral hemorrhage. Effects of intra-arterial iodinated radiographic contrast material on hemorrhagic transformation after middle cerebral artery occlusion and reperfusion were studied in a placebo-controlled, blinded preclinical study in rats. METHODS: Four groups of male Sprague-Dawley rats were studied: saline group (n=8), contrast group (n=12), heparin group (n=9), and contrast+heparin group (n=9). The middle cerebral artery was occluded for 5 hours using suture placement. Heparin was infused before suture removal and reperfusion. Saline and/or contrast were infused immediately during reperfusion. Incidence, location, and size of hemorrhage were determined by brain necropsy inspection at 24 hours. RESULTS: There was a significant increase in incidence of cortical hemorrhage from control (37.5%), contrast (75.0%), heparin (77.8%) to contrast+heparin (100%; Cochran-Mantel-Haenszel correlation, P<0.01). Both pooled contrast groups (85.7%) and pooled heparin groups (88.9%) had higher rates of cortical intracerebral hemorrhage compared with the control group (P<0.05). Similar trends for increased cortical intracerebral hemorrhage were seen in the contrast-only (P=0.18) and heparin-only (P=0.18) groups. There was a trend for decreased infarct edema in rats receiving contrast versus those without (P=0.06). CONCLUSIONS: Intraarterial iodinated radiographic contrast material may increase cortical intracerebral hemorrhage, similar to heparin. Iodinated radiographic contrast material effect may be additive to heparin effect on the incidence of cortical intracerebral hemorrhage.


Subject(s)
Cerebral Hemorrhage/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Iodine Radioisotopes/adverse effects , Iohexol/adverse effects , Reperfusion Injury/pathology , Animals , Cerebral Hemorrhage/chemically induced , Contrast Media/administration & dosage , Contrast Media/adverse effects , Infarction, Middle Cerebral Artery/diagnosis , Injections, Intra-Arterial , Iodine Radioisotopes/administration & dosage , Iohexol/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/diagnosis , Single-Blind Method
9.
Huan Jing Ke Xue ; 30(2): 362-7, 2009 Feb 15.
Article in Chinese | MEDLINE | ID: mdl-19402482

ABSTRACT

Rainwater samples were colleted from Lijiang City, China, in 23 May-2 July, 2006. Rainwater chemical compositions and sources were studied, using HYSPLIT model, ions tracer techniques, correlation and trend analysis. Total ionic concentration was dominated by SO4(2-) and Ca2+, which account for 65.5% and 15.6% respectively. Sort order of ions concentration is SO4(2-) > Ca2+ > Cl(-) > NO3(-) > Na+ > K+ > Mg2+. Total anions concentration is higher than total cations concentration in 13 rainwater events. The ratio of SO4(2-) to NO3(-) varies from 7.2 to 37.1 and average value is 15.7, it reflected SO4(2-) made great contribution to rainwater acidity in Lijiang City. The correlation among ions is significant due to the atmospheric chemical process and similar ionic sources, and correlation coefficient between SO4(2-) and NO3(-) is 0.74. And what's more, the negative correlation of ionic concentration, precipitation and the average wind speed is also outstanding. The source of NO3(-), SO4(2-), K+ and Ca2+ is mainly land dust, and the non-marine source percent of NO3(-), SO4(2-), K+, Ca2+, Mg2+ and Cl(-) is 100%, 98.8%, 96%, 99.3%, 46.7% and 50.3%, respectively. The main reason of atmospheric environmental variation in Lijing City is pollution caused by economic actions. The pollutants from surrounding industrial parks input into Lijiang City by local circulation, and from industrial regions of Southern Asia, Southeastern Asia and Southeastern China input into Lijiang City by monsoonal circulation.


Subject(s)
Acid Rain/analysis , Air Pollutants/analysis , Nitrates/analysis , Rain/chemistry , Sulfur Dioxide/analysis , Calcium/analysis , China , Seasons
10.
Neurol Res ; 31(2): 209-14, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19298764

ABSTRACT

OBJECTIVE: Recanalization therapies for ischemic stroke have been slow to change clinical practice because of perceived and published risks of hemorrhage associated with lytic administration. We quantified alfimeprase in an acute ischemia-reperfusion model, as compared with recombinant tissue plasminogen activator, with hemorrhagic transformation as the primary endpoint and infarction volume and blood-brain barrier permeability as secondary endpoints. METHODS: Five groups were studied in a blinded fashion: alfimeprase at doses of 0.03 (n=8), 0.1 (n=11) and 0.3 mg/kg (n=8); recombinant tissue plasminogen activator at 1 mg/kg (n=9); carrier infused controls (n=9). The middle cerebral artery was occluded for 5 hours followed by removal of the suture for reperfusion. Drugs were infused immediately following reperfusion over a 10-minute period. Approximately 24 hours later, the animals were anesthetized and decapitated, and the brains were rapidly harvested and frozen. Serial brain sections were obtained and inspected for hemorrhages. Infarction and blood-brain barrier permeability were also evaluated in additional experiments in control, 0.1 mg/kg alfimeprase and 1 mg/kg recombinant tissue plasminogen activator-treated rats. RESULTS: The hemorrhagic transformation frequency, neurological deficit and the mortality rate of alfimeprase were significantly lower than for recombinant tissue plasminogen activator at the 0.03 mg/kg dose and not statistically different at the higher doses. Infarction and blood-brain barrier permeability were not significantly different among control, 0.1 mg/kg alfimeprase and recombinant tissue plasminogen activator. DISCUSSION: In this model, alfimeprase, a new fibrinolytic agent, exhibits a profile comparable to recombinant tissue plasminogen activator.


Subject(s)
Cerebral Hemorrhage/chemically induced , Fibrinolytic Agents/adverse effects , Infarction, Middle Cerebral Artery/drug therapy , Metalloendopeptidases/adverse effects , Reperfusion Injury/chemically induced , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Body Weight/drug effects , Brain Edema/drug therapy , Brain Edema/pathology , Capillary Permeability/drug effects , Cerebral Hemorrhage/mortality , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Chi-Square Distribution , Disease Models, Animal , Dose-Response Relationship, Drug , Double-Blind Method , Fibrinolytic Agents/therapeutic use , Infarction, Middle Cerebral Artery/mortality , Male , Metalloendopeptidases/drug effects , Neurologic Examination , Rats , Reperfusion Injury/mortality , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Time Factors , Tissue Plasminogen Activator/administration & dosage
11.
Exp Neurol ; 216(2): 407-12, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19162014

ABSTRACT

A major complication of recanalization therapy after an acute arterial occlusion in brain is hemorrhagic transformation (HT). Although it is known that prolonged ischemia is important in the development of HT, the role of reperfusion in ischemia-reperfusion induced HT is less well studied. To address the effect of reperfusion on HT, we assessed the incidence and severity of hemorrhage in rats after 5 h of middle cerebral artery occlusion (MCAO) followed by 19-hour reperfusion compared to rats with permanent occlusion (PMCAO) at the same 24-hour time point. The incidence and amount of hemorrhage, neurological function, and mortality rates were measured. MCAO (5 h) with 19-hour reperfusion was associated with a significantly higher incidence of cortical hemorrhage compared to PMCAO (81.8% vs 18.2%, p<0.05). Hemorrhage scores were higher in the 5-hour MCAO/reperfusion group compared to PMCAO rats (17.6+/-11.5 vs 2.4+/-5.3 in cortex, 20.4+/-4.6 vs 9.7+/-4.5 in striatum, p<0.01). Neurological function was worse in the ischemia-reperfusion group compared to PMCAO (p<0.05) and mortality rates were insignificantly higher in the 5-hour MCAO/reperfusion group vs PMCAO group (54.5% vs 18.1%; p<0.08). The results suggest that reperfusion after prolonged ischemia is associated with increased hemorrhagic transformation and neurological deterioration as compared to permanent ischemia. Whether pharmacological treatments prior to reperfusion attenuate post-ischemic HT requires further study.


Subject(s)
Cerebral Hemorrhage/etiology , Infarction, Middle Cerebral Artery/physiopathology , Reperfusion/adverse effects , Analysis of Variance , Animals , Behavior, Animal , Brain/pathology , Brain Edema/etiology , Brain Edema/pathology , Brain Infarction/etiology , Brain Infarction/pathology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/therapy , Male , Nervous System Diseases/etiology , Nervous System Diseases/pathology , Rats , Rats, Sprague-Dawley , Time Factors
12.
J Cereb Blood Flow Metab ; 29(1): 137-43, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18827833

ABSTRACT

Intracerebral hemorrhage (ICH) is a stroke subtype with high rates of mortality and morbidity. The immune system, particularly complement and cytokine signaling, has been implicated in brain injury after ICH. However, the cellular immunology associated with ICH has been understudied. In this report, we use flow cytometry to quantitatively profile immune cell populations that infiltrate the brain 1 and 4 days post-ICH. At 1 day CD45(hi) GR-1(+) cells were increased 2.0-fold compared with saline controls (P

Subject(s)
Cell Movement , Cerebral Hemorrhage/physiopathology , Leukocytes/cytology , Animals , Behavior, Animal , Cerebral Hemorrhage/metabolism , Leukocyte Common Antigens/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL
13.
Neurol Res ; 31(5): 545-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19055876

ABSTRACT

OBJECTIVE: The discovery of IL-7R(alpha) polymorphisms implicated in the pathogenesis of multiple sclerosis has highlighted the importance of interleukin 7 (IL-7) in central nervous system diseases. Hypoxia affects neurological disease states in part by modulating expression of many early and late response genes. The present work used cultured PC12 cells to investigate the effect of hypoxia on IL-7 expression. METHOD: PC12 cells were cultured in Dulbecco's modified Eagle's medium (DMEM)/F12 medium. RNA was isolated and reverse transcriptase-polymerase chain reaction (RT-PCR) was run to quantify messenger RNA (mRNA) change. Western blots were used to assess IL-7 protein change in the medium. Extracellular free Ca(2+) was removed by using Ca(2+)-free DMEM/F12 with 1 mM ethylene glycol tetraacetic acid for 45 minutes before the start of hypoxia. RESULTS: Exposure of PC12 cells to 1% oxygen for 6 hours decreased IL-7 mRNA by 77% using RT-PCR (p<0.01). Exposure to 1% oxygen for 24 hours decreased IL-7 protein in the medium by 21% (p<0.05). As hypoxia duration increased (2, 4, 6 and 24 hours) or oxygen concentrations decreased (10%, 5% and 1%), IL-7 mRNA expression progressively decreased. Removal of extracellular free Ca(2+) completely prevented these hypoxia-induced decreases of IL-7 mRNA. DISCUSSION: Since IL-7 exhibits trophic properties in developing brain, down-regulation of IL-7 by hypoxia may contribute to hypoxia-induced injury to neural cells.


Subject(s)
Calcium/pharmacology , Cell Hypoxia/genetics , Interleukin-7/metabolism , Animals , Blotting, Western , Down-Regulation , Oxygen/pharmacology , PC12 Cells , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
14.
Exp Neurol ; 210(2): 549-59, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18187134

ABSTRACT

In this study, we examine the effects of reperfusion on the activation of matrix metalloproteinase (MMP) and assess the relationship between MMP activation during reperfusion and neurovascular injury. Ischemia was produced using suture-induced middle cerebral artery occlusion in rats. The MMP activation was examined with in situ and gel zymography. Injury to cerebral endothelial cells and basal lamina was assessed using endothelial barrier antigen (EBA) and collagen IV immunohistochemistry. Injury to neurons and glial cells was assessed using Cresyl violet staining. These were examined at 3 h after reperfusion (8 h after initiation of ischemia) and compared with permanent ischemia at the same time points to assess the effects of reperfusion. A broad-spectrum MMP inhibitor, AHA (p-aminobenzoyl-Gly-Pro-D-Leu-D-Ala-hydroxamate, 50 mg/kg intravenously) was administered 30 min before reperfusion to assess the roles of MMPs in activating gelatinolytic enzymes and in reperfusion-induced injury. We found that reperfusion accelerated and potentiated MMP-9 and MMP-2 activation and injury to EBA and collagen IV immunopositive microvasculature and to neurons and glial cells in ischemic cortex and striatum relative to permanent ischemia. Administering AHA 30 min before reperfusion decreased MMP-9 activation and neurovascular injury in ischemic cerebral cortex.


Subject(s)
Basement Membrane/enzymology , Cerebrovascular Trauma/etiology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Reperfusion Injury/enzymology , Reperfusion/adverse effects , Analysis of Variance , Animals , Autoantigens/metabolism , Basement Membrane/drug effects , Basement Membrane/pathology , Collagen Type IV/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Time Factors
15.
Mol Cell Neurosci ; 36(3): 392-407, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17888676

ABSTRACT

A cDNA encoding a novel protein was cloned from ischemic rat brain and found to be homologous to testis Mea-2 Golgi-associated protein (Golga3). The sequence predicted a 165-kDa protein, and in vitro translated protein exhibited a molecular mass of 165-170 kDa. Because brain ischemia induced the mRNA, and the protein localized to the Golgi apparatus, this protein was designated Ischemia-Inducible Golgin Protein 165 (IIGP165). In HeLa cells, serum and glucose deprivation-induced caspase-dependent cleavage of the IIGP165 protein, after which the IIGP165 fragments translocated to the nucleus. The C-terminus of IIGP165, which contains a LXXLL motif, appears to function as a transcriptional co-regulator. Akt co-localizes with IIGP165 protein in the Golgi in vivo, and phosphorylates IIGP165 on serine residues 345 and 134. Though transfection of IIGP165 cDNA alone does not protect HeLa cells from serum deprivation or Brefeldin-A-triggered cell death, co-transfection of both Akt and IIGP165 cDNA or combined IIGP165-transfection with PDGF treatment significantly protects HeLa cells better than either treatment alone. These data show that Akt phosphorylation of IIGP165 protects against apoptotic cell death, and add to evidence that the Golgi apparatus also plays a role in regulating apoptosis.


Subject(s)
Apoptosis/physiology , Brain Ischemia/metabolism , Brain/metabolism , Cytoprotection/genetics , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence/physiology , Animals , Autoantigens/chemistry , Autoantigens/genetics , Autoantigens/metabolism , Base Sequence , Brain/physiopathology , Brain Ischemia/genetics , Brain Ischemia/physiopathology , COS Cells , Chlorocebus aethiops , Cytoprotection/drug effects , DNA, Complementary/pharmacology , Golgi Matrix Proteins , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Molecular Sequence Data , PC12 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/isolation & purification , Transfection/methods
16.
Med Hypotheses ; 67(3): 506-12, 2006.
Article in English | MEDLINE | ID: mdl-16730130

ABSTRACT

N-acetylaspartate (NAA) is an intermediary metabolite that is found in relatively high concentrations in the human brain. More specifically, NAA is so concentrated in the neurons that it generates one of the most visible peaks in nuclear magnetic resonance (NMR) spectra, thus allowing NAA to serve as "a neuronal marker". However, to date there is no generally accepted physiological (primary) role for NAA. Another molecule that is found at similar concentrations in the brain is glutamate. Glutamate is an amino acid and neurotransmitter with numerous functions in the brain. We propose that NAA, a six-carbon amino acid derivative, is converted to glutamate (five carbons) in an energetically favorable set of reactions. This set of reactions starts when aspartoacylase converts the six carbons of NAA to aspartate and acetate, which are subsequently converted to oxaloacetate and acetyl CoA, respectively. Aspartylacylase is found in astrocytes and oligodendrocytes. In the mitochondria, oxaloacetate and acetyl CoA are combined to form citrate. Requiring two steps, the citrate is oxidized in the Kreb's cycle to alpha-ketoglutarate, producing NADH. Finally, alpha-ketoglutarate is readily converted to glutamate by transaminating the alpha-keto to an amine. The resulting glutamate can be used by multiple cells types to provide optimal brain functional and structural needs. Thus, the abundant NAA in neuronal tissue can serve as a large reservoir for replenishing glutamate in times of rapid or dynamic signaling demands and stress. This is beneficial in that proper levels of glutamate serve critical functions for neurons, astrocytes, and oligodendrocytes including their survival. In conclusion, we hypothesize that NAA conversion to glutamate is a logical and favorable use of this highly concentrated metabolite. It is important for normal brain function because of the brain's relatively unique metabolic demands and metabolite fluxes. Knowing that NAA is converted to glutamate will be important for better understanding myriad neurodegenerative diseases such as Canavan's Disease and Multiple Sclerosis, to name a few. Future studies to demonstrate the chemical, metabolic and pathological links between NAA and glutamate will support this hypothesis.


Subject(s)
Aspartic Acid/analogs & derivatives , Aspartic Acid/physiology , Dipeptides/physiology , Glutamic Acid/physiology , Aspartic Acid/chemistry , Astrocytes/physiology , Brain/physiology , Citric Acid Cycle , Glutamic Acid/chemistry , Humans , Models, Biological , Neurons/physiology , Neurotransmitter Agents/physiology , Oligodendroglia/physiology
17.
J Cereb Blood Flow Metab ; 26(8): 1089-102, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16395289

ABSTRACT

Ischemic brain and peripheral white blood cells release cytokines, chemokines and other molecules that activate the peripheral white blood cells after stroke. To assess gene expression in these peripheral white blood cells, whole blood was examined using oligonucleotide microarrays in 15 patients at 2.4+/-0.5, 5 and 24 h after onset of ischemic stroke and compared with control blood samples. The 2.4-h blood samples were drawn before patients were treated either with tissue-type plasminogen activator (tPA) alone or with tPA plus Eptifibatide (the Combination approach to Lysis utilizing Eptifibatide And Recombinant tPA trial). Most genes induced in whole blood at 2 to 3 h were also induced at 5 and 24 h. Separate studies showed that the genes induced at 2 to 24 h after stroke were expressed mainly by polymorphonuclear leukocytes and to a lesser degree by monocytes. These genes included: matrix metalloproteinase 9; S100 calcium-binding proteins P, A12 and A9; coagulation factor V; arginase I; carbonic anhydrase IV; lymphocyte antigen 96 (cluster of differentiation (CD)96); monocarboxylic acid transporter (6); ets-2 (erythroblastosis virus E26 oncogene homolog 2); homeobox gene Hox 1.11; cytoskeleton-associated protein 4; N-formylpeptide receptor; ribonuclease-2; N-acetylneuraminate pyruvate lyase; BCL6; glycogen phosphorylase. The fold change of these genes varied from 1.6 to 6.8 and these 18 genes correctly classified 10/15 patients at 2.4 h, 13/15 patients at 5 h and 15/15 patients at 24 h after stroke. These data provide insights into the inflammatory responses after stroke in humans, and should be helpful in diagnosis, understanding etiology and pathogenesis, and guiding acute treatment and development of new treatments for stroke.


Subject(s)
Brain Ischemia/blood , Gene Expression Regulation , Monocytes/metabolism , Neutrophils/metabolism , Stroke/blood , Adult , Aged , Brain Ischemia/drug therapy , Drug Therapy, Combination , Eptifibatide , Female , Fibrinolytic Agents/therapeutic use , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Inflammation/blood , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Peptides/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Stroke/drug therapy , Time Factors , Tissue Plasminogen Activator/therapeutic use
18.
J Cereb Blood Flow Metab ; 26(2): 230-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16034371

ABSTRACT

After intracerebral hemorrhage (ICH), many changes of gene transcription occur that may be important because they will contribute to understanding mechanisms of injury and recovery. Therefore, gene expression was assessed using Affymetrix microarrays in the striatum and the overlying cortex at 24 h after intracranial infusions of blood into the striatum of adult rats. Intracerebral hemorrhage regulated 369 of 8,740 transcripts as compared with saline-injected controls, with 104 regulated genes shared by the striatum and cortex. There were 108 upregulated and 126 downregulated genes in striatum, and 170 upregulated and 69 downregulated genes in the cortex. Real-time reverse transcriptase-polymerase chain reaction (RT-PCR) confirmed upregulation of IL-1-beta, Lipcortin 1 (annexin) and metallothionein 1,2, and downregulation of potassium voltage-gated channel, shaker-related subfamily, beta member 2 (Kcnab2). Of the functional groups of genes modulated by ICH, many metabolism and signal-transduction-related genes decreased in striatum but increased in adjacent cortex. In contrast, most enzyme, cytokine, chemokine, and immune response genes were upregulated in both striatum and in the cortex after ICH, likely in response to foreign proteins from the blood. A number of these genes may contribute to brain edema and cellular apoptosis caused by ICH. In addition, downregulation of growth factor pathways and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway could also contribute to perihematoma cell death/apoptosis. Intracerebral hemorrhage-related downregulation of GABA-related genes and potassium channels might contribute to perihematoma cellular excitability and increased risk of post-ICH seizures. These genomic responses to ICH potentially provide new therapeutic targets for treatment.


Subject(s)
Brain/physiology , Cerebral Hemorrhage/genetics , Gene Expression Profiling , Genomics , Animals , Annexin A1/genetics , Annexin A1/metabolism , Brain/metabolism , Cluster Analysis , Disease Models, Animal , Down-Regulation , Growth Substances/genetics , Growth Substances/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Male , Metallothionein/genetics , Metallothionein/metabolism , Oligonucleotide Array Sequence Analysis/methods , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic , Up-Regulation , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
19.
Dev Neurosci ; 27(2-4): 87-92, 2005.
Article in English | MEDLINE | ID: mdl-16046841

ABSTRACT

Exposure to moderate hypoxia alone does not cause neuronal death as long as blood pressure and cerebral blood flow are maintained in mammals. In neonatal and adult mammals including rats and mice, carotid occlusion in combination with hypoxia produces neuronal death and brain infarction. However, preexposure to 8% oxygen for 3 h protects the brain and likely other organs of neonatal and adult rats against combined hypoxia-ischemia 24 h later. In this paper, the possible mechanisms of this so-called hypoxia-induced tolerance to ischemia is discussed. One mechanism likely involves hypoxia-inducible factor-1alpha (HIF-1alpha). HIF-1alpha is a transcription factor that - during hypoxia - binds with a second protein (HIF-1beta) in the nucleus to promoter elements in hypoxia-responsive target genes. This causes upregulation of HIF target genes including VEGF, erythropoietin, iNOS, glucose transporter-1, glycolytic enzymes, and many other genes to protect the brain against ischemia 24 h later. In addition, non-HIF pathways including MTF-1, Egr-1 and others act directly or indirectly on other target genes to also promote hypoxia-induced preconditioning. Hypoxia preconditioning can be mimicked by iron chelators like desferrioxamine and transition metals like cobalt chloride that inhibit prolyl hydroxylases, increase HIF-1alpha levels in the brain, and produce protection of the brain against combined hypoxia-ischemia 24 h later. This hypoxia preconditioning has potential clinical usefulness in protecting high-risk newborns or to provide protection prior to surgery.


Subject(s)
Brain/blood supply , Hypoxia-Ischemia, Brain/physiopathology , Ischemic Preconditioning , Models, Biological , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Rats , Transcription Factors/metabolism
20.
Brain Res Mol Brain Res ; 132(2): 155-67, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15582155

ABSTRACT

Application of gene expression profiling to human diseases will be limited by availability of tissue samples. It was postulated that germline genetic defects affect blood cells to produce unique expression patterns. This hypothesis was addressed by using a test neurological disease-neurofibromatosis type 1 (NF1), an autosomal dominant genetic disease caused by mutations of the NF1 gene at chromosome 17q11.2. Oligonucleotide arrays were used to survey the blood gene expression pattern of 12 NF1 patients compared to 96 controls. A group of genes related to tissue remodeling, bone development and tumor suppression were down-regulated in NF1 blood samples. In addition, there were blood genomic patterns for gender and age: Y chromosome genes showing higher expression in males, indicating a gene-dosage effect; and genes related to lymphocyte functions showing higher expression in children. The results suggest that genetic mutations can be manifested at the transcriptional level in peripheral blood cells and blood gene expression profiling may be useful for studying phenotypic differences of human genetic diseases and possibly providing diagnostic and prognostic markers.


Subject(s)
Blood Physiological Phenomena , Genomics , Neurofibromatosis 1/blood , Neurofibromatosis 1/genetics , Adolescent , Adult , Age Factors , Female , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...