Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(714): eadg8656, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37729432

ABSTRACT

Human diseases may be modeled in animals to allow preclinical assessment of putative new clinical interventions. Recent, highly publicized failures of large clinical trials called into question the rigor, design, and value of preclinical assessment. We established the Stroke Preclinical Assessment Network (SPAN) to design and implement a randomized, controlled, blinded, multi-laboratory trial for the rigorous assessment of candidate stroke treatments combined with intravascular thrombectomy. Efficacy and futility boundaries in a multi-arm multi-stage statistical design aimed to exclude from further study highly effective or futile interventions after each of four sequential stages. Six independent research laboratories performed a standard focal cerebral ischemic insult in five animal models that included equal numbers of males and females: young mice, young rats, aging mice, mice with diet-induced obesity, and spontaneously hypertensive rats. The laboratories adhered to a common protocol and efficiently enrolled 2615 animals with full data completion and comprehensive animal tracking. SPAN successfully implemented treatment masking, randomization, prerandomization inclusion and exclusion criteria, and blinded assessment of outcomes. The SPAN design and infrastructure provide an effective approach that could be used in similar preclinical, multi-laboratory studies in other disease areas and should help improve reproducibility in translational science.


Subject(s)
Ischemic Stroke , Stroke , Female , Humans , Male , Rats , Animals , Mice , Rodentia , Laboratories , Reproducibility of Results , Stroke/therapy
2.
J Cereb Blood Flow Metab ; 43(11): 1842-1856, 2023 11.
Article in English | MEDLINE | ID: mdl-37466218

ABSTRACT

Neurologic outcome from out-of-hospital pediatric cardiac arrest remains poor. Although therapeutic hypothermia has been attempted in this patient population, a beneficial effect has yet to be demonstrated, possibly because of the delay in achieving target temperature. To minimize this delay, we developed a simple technique of transnasal cooling. Air at ambient temperature is passed through standard nasal cannula with an open mouth to produce evaporative cooling of the nasal passages. We evaluated efficacy of brain cooling with different airflows in different size piglets. Brain temperature decreased by 3°C within 25 minutes with nasal airflow rates of 16, 32, and 16 L/min in 1.8-, 4-, and 15-kg piglets, respectively, whereas rectal temperature lagged brain temperature. No substantial spatial temperature gradients were seen along the neuroaxis, suggesting that heat transfer is via blood convection. The evaporative cooling did not reduce nasal turbinate blood flow or sagittal sinus oxygenation. The rapid and selective brain cooling indicates a high humidifying capacity of the nasal turbinates is present early in life. Because of its simplicity, portability, and low cost, transnasal cooling potentially could be deployed in the field for early initiation of brain cooling prior to maintenance with standard surface cooling after pediatric cardiac arrest.


Subject(s)
Cardiopulmonary Resuscitation , Hypothermia, Induced , Out-of-Hospital Cardiac Arrest , Humans , Animals , Child , Swine , Hypothermia, Induced/methods , Cold Temperature , Body Temperature/physiology , Brain , Cardiopulmonary Resuscitation/methods
3.
Front Med Technol ; 5: 1074643, 2023.
Article in English | MEDLINE | ID: mdl-36896342

ABSTRACT

Polynitroxylated PEGylated hemoglobin (PNPH, aka SanFlow) possesses superoxide dismutase/catalase mimetic activities that may directly protect the brain from oxidative stress. Stabilization of PNPH with bound carbon monoxide prevents methemoglobin formation during storage and permits it to serve as an anti-inflammatory carbon monoxide donor. We determined whether small volume transfusion of hyperoncotic PNPH is neuroprotective in a porcine model of traumatic brain injury (TBI) with and without accompanying hemorrhagic shock (HS). TBI was produced by controlled cortical impact over the frontal lobe of anesthetized juvenile pigs. Hemorrhagic shock was induced starting 5 min after TBI by 30 ml/kg blood withdrawal. At 120 min after TBI, pigs were resuscitated with 60 ml/kg lactated Ringer's (LR) or 10 or 20 ml/kg PNPH. Mean arterial pressure recovered to approximately 100 mmHg in all groups. A significant amount of PNPH was retained in the plasma over the first day of recovery. At 4 days of recovery in the LR-resuscitated group, the volume of frontal lobe subcortical white matter ipsilateral to the injury was 26.2 ± 7.6% smaller than homotypic contralateral volume, whereas this white matter loss was only 8.6 ± 12.0% with 20-ml/kg PNPH resuscitation. Amyloid precursor protein punctate accumulation, a marker of axonopathy, increased in ipsilateral subcortical white matter by 132 ± 71% after LR resuscitation, whereas the changes after 10 ml/kg (36 ± 41%) and 20 ml/kg (26 ± 15%) PNPH resuscitation were not significantly different from controls. The number of cortical neuron long dendrites enriched in microtubules (length >50 microns) decreased in neocortex by 41 ± 24% after LR resuscitation but was not significantly changed after PNPH resuscitation. The perilesion microglia density increased by 45 ± 24% after LR resuscitation but was unchanged after 20 ml/kg PNPH resuscitation (4 ± 18%). Furthermore, the number with an activated morphology was attenuated by 30 ± 10%. In TBI pigs without HS followed 2 h later by infusion of 10 ml/kg LR or PNPH, PNPH remained neuroprotective. These results in a gyrencephalic brain show that resuscitation from TBI + HS with PNPH protects neocortical gray matter, including dendritic microstructure, and white matter axons and myelin. This neuroprotective effect persists with TBI alone, indicating brain-targeting benefits independent of blood pressure restoration.

4.
J Neural Eng ; 18(6)2022 01 05.
Article in English | MEDLINE | ID: mdl-34937013

ABSTRACT

OBJECTIVE: Perinatal ischemic stroke is estimated to occur in 1/2300-1/5000 live births, but early differential diagnosis from global hypoxia-ischemia is often difficult. In this study, we tested the ability of a hand-held transcranial photoacoustic (PA) imaging probe to non-invasively detect a focal photothrombotic stroke (PTS) within 2 h of stroke onset in a gyrencephalic piglet brain. APPROACH: About 17 stroke lesions of approximately 1 cm2area were introduced randomly in anterior or posterior cortex via the light/dye PTS technique in anesthetized neonatal piglets (n= 11). The contralateral non-ischemic region served as control tissue for discrimination contrast for the PA hemoglobin metrics: oxygen saturation, total hemoglobin (tHb), and individual quantities of oxygenated and deoxygenated hemoglobin (HbO2and HbR). MAIN RESULTS: The PA-derived tissue oxygen saturation at 2 h yielded a significant separation between control and affected regions-of-interest (p< 0.0001), which were well matched with 24 h post-stroke cerebral infarction confirmed in the triphenyltetrazolium chloride-stained image. The quantity of HbO2also displayed a significant contrast (p= 0.021), whereas tHb and HbR did not. The analysis on receiver operating characteristic curves and multivariate data analysis also agreed with the results above. SIGNIFICANCE: This study shows that a hand-held transcranial PA neuroimaging device can detect a regional thrombotic stroke in the cerebral cortex of a neonatal piglet. In particular, we conclude that the oxygen saturation metric can be used alone to identify regional stroke lesions. The lack of change in tHb may be related to arbitrary hand-held imaging configuration and/or entrapment of red blood cells within the thrombotic stroke.


Subject(s)
Brain Ischemia , Stroke , Animals , Brain , Brain Ischemia/pathology , Cerebral Cortex , Neuroimaging , Stroke/diagnostic imaging , Swine
5.
Magn Reson Med ; 84(6): 3342-3350, 2020 12.
Article in English | MEDLINE | ID: mdl-32597519

ABSTRACT

PURPOSE: To obtain high-sensitivity CEST maps by exploiting the spatiotemporal correlation between CEST images. METHODS: A postprocessing method accomplished by multilinear singular value decomposition (MLSVD) was used to enhance the CEST SNR by exploiting the correlation between the Z-spectrum for each voxel and the low-rank property of the overall CEST data. The performance of this method was evaluated using CrCEST in ischemic mouse brain at 11.7 tesla. Then, MLSVD CEST was applied to obtain Cr, amide, and amine CEST maps of the ischemic mouse brain to demonstrate its general applications. RESULTS: Complex-valued Gaussian noise was added to CEST k-space data to mimic a low SNR situation. MLSVD CEST analysis was able to suppress the noise, recover the degraded CEST peak, and provide better CrCEST quality compared to the smoothing and singular value decomposition (SVD)-based denoising methods. High-resolution Cr, amide, and amine CEST maps of an ischemic stroke using MLSVD CEST suggest that CrCEST is also a sensitive pH mapping method, and a wide range of pH changes can be detected by combing CrCEST with amine CEST at high magnetic fields. CONCLUSION: MLSVD CEST provides a simple and efficient way to improve the SNR of CEST images.


Subject(s)
Amides , Magnetic Resonance Imaging , Animals , Mice
6.
BMC Neurosci ; 21(1): 22, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32404052

ABSTRACT

BACKGROUND: Polynitroxylated PEGylated hemoglobin (PNPH, aka SanFlow) possesses superoxide dismutase/catalase mimetic activities that may directly protect the brain from oxidative stress. Stabilization of PNPH with bound carbon monoxide prevents methemoglobin formation during storage and permits it to serve as a carbon monoxide donor. We determined whether small volume transfusion of hyperoncotic PNPH is neuroprotective in a polytrauma model of traumatic brain injury (TBI) plus hemorrhagic shock. Guinea pigs were used because, like humans, they do not synthesize their own ascorbic acid, which is important in reducing methemoglobin. RESULTS: TBI was produced by controlled cortical impact and was followed by 20 mL/kg hemorrhage to a mean arterial pressure (MAP) of 40 mmHg. At 90 min, animals were resuscitated with 20 mL/kg lactated Ringer's solution or 10 mL/kg PNPH. Resuscitation with PNPH significantly augmented the early recovery of MAP after hemorrhagic shock by 10-18 mmHg; whole blood methemoglobin was only 1% higher and carboxyhemoglobin was 2% higher. At 9 days of recovery, unbiased stereology analysis revealed that, compared to animals resuscitated with lactated Ringer's solution, those treated with PNPH had significantly more viable neurons in the hippocampus CA1 + 2 region (59 ± 10% versus 87 ± 18% of sham and naïve mean value) and in the dentate gyrus (70 ± 21% versus 96 ± 24%; n = 12 per group). CONCLUSION: PNPH may serve as a small-volume resuscitation fluid for polytrauma involving TBI and hemorrhagic shock. The neuroprotection afforded by PNPH seen in other species was sustained in a species without endogenous ascorbic acid synthesis, thereby supporting potential translatability for human use.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Catalase/pharmacology , Hemorrhage/drug therapy , Resuscitation , Shock, Hemorrhagic/drug therapy , Animals , Guinea Pigs , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology
7.
J Cereb Blood Flow Metab ; 38(5): 835-846, 2018 05.
Article in English | MEDLINE | ID: mdl-28436294

ABSTRACT

The loss of oligodendrocytes after stroke is one of the major causes of secondary injury. Glial-restricted progenitors (GRPs) have remylenating potential after intraparenchymal cerebral transplantation. The intraarterial (IA) injection route is an attractive gateway for global brain delivery, but, after IA infusion, naive GRPs fail to bind to the cerebral vasculature. The aim of this study was to test whether overexpression of Very Late Antigen-4 (VLA-4) increases endothelial docking and cerebral homing of GRPs in a stroke model. Mouse GRPs were co-transfected with DNA plasmids encoding VLA-4 subunits (α4, ß1). The adhesion capacity and migration were assessed using a microfluidic assay. In vivo imaging of the docking and homing of IA-infused cells was performed using two-photon microscopy in a mouse middle cerebral artery occlusion (MCAO) model. Compared to naïve GRPs, transfection of GRPs with VLA-4 resulted in >60% higher adhesion (p < 0.05) to both purified Vascular Cell Adhesion Molecule-11 (VCAM-11) and TNFα-induced endothelial VCAM-1. VLA-4+GRPs displayed a higher migration in response to a chemoattractant gradient. Following IA infusion, VLA-4+GRPs adhered to the vasculature at three-fold greater numbers than naïve GRPs. Multi-photon imaging confirmed that VLA-4 overexpression increases the efficiency of GRP docking and leads to diapedesis after IA transplantation. This strategy may be further exploited to increase the efficacy of cellular therapeutics.


Subject(s)
Integrin alpha4beta1/metabolism , Neural Stem Cells/transplantation , Neuroglia/transplantation , Stroke , Transendothelial and Transepithelial Migration/physiology , Animals , Blood-Brain Barrier , Cell Adhesion/physiology , Cell Movement/physiology , Disease Models, Animal , Endothelium, Vascular/metabolism , Mice , Neural Stem Cells/metabolism , Neuroglia/metabolism , Stem Cell Transplantation/methods
8.
J Am Heart Assoc ; 6(9)2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28899897

ABSTRACT

BACKGROUND: Polynitroxylation of hemoglobin confers superoxide dismutase-mimetic and peroxidase activity and may protect from reperfusion injury in addition to facilitating oxygen transport. We determined whether transfusion of polynitroxylated PEGylated hemoglobin (PNPH) is protective in the rat filament model of 2 hours of middle cerebral artery occlusion (MCAO). METHODS AND RESULTS: Transfusion of 10 mL/kg of PNPH at 20 minutes of MCAO reduced infarct volume by over 70% (n=10). To determine whether PNPH might act by promoting vasodilation, pial arteriolar diameter in the distal MCA border region was measured in closed cranial windows. With no transfusion, MCAO induced an initial dilation (36±2% ±SE) that subsided by 2 hours (5±4%; n=8). With PNPH transfusion at 20 minutes of MCAO, the initial dilation (31±3%) was better maintained at 2 hours (21±4%; n=7; P<0.02). Delaying PNPH transfusion until 90 minutes of MCAO increased perfusion in the border region from 48±6% of the preischemic baseline to 67±8% (n=8; P<0.005). The effect of PNPH transfusion after reperfusion was also tested. Compared with the control median hemispheric infarct volume of 22% (13% to 34% interquartiles; n=15), infarct volume was reduced to 7% (3% to 13%; n=14 P<0.05) when PNPH was transfused at 4 hours after MCAO (2 hours of reperfusion) but not significantly when transfused at 6 hours (8%; 3% to 35%; n=14) or at 8 hours (12%; 10% to 25%; n=14) after MCAO. CONCLUSIONS: PNPH transfusion has a significant therapeutic window for protection during and after transient MCAO and may act, in part, by stabilizing vascular function and improving collateral blood flow.


Subject(s)
Cerebral Arteries/drug effects , Cerebrovascular Circulation/drug effects , Hemoglobins/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/administration & dosage , Pia Mater/blood supply , Reperfusion Injury/prevention & control , Vasodilation/drug effects , Animals , Cerebral Arteries/physiopathology , Collateral Circulation/drug effects , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Infusions, Intravenous , Male , Rats, Wistar , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Time Factors
9.
Theranostics ; 6(10): 1588-600, 2016.
Article in English | MEDLINE | ID: mdl-27446492

ABSTRACT

Citicoline (CDPC) is a natural supplement with well-documented neuroprotective effects in the treatment of neurodegenerative diseases. In the present study, we sought to exploit citicoline as a theranostic agent with its inherent chemical exchange saturation transfer (CEST) MRI signal, which can be directly used as an MRI guidance in the citicoline drug delivery. Our in vitro CEST MRI results showed citicoline has two inherent CEST signals at +1 and +2 ppm, attributed to exchangeable hydroxyl and amine protons, respectively. To facilitate the targeted drug delivery of citicoline to ischemic regions, we prepared liposomes encapsulating citicoline (CDPC-lipo) and characterized the particle properties and CEST MRI properties. The in vivo CEST MRI detection of liposomal citicoline was then examined in a rat brain model of unilateral transient ischemia induced by a two-hour middle cerebral artery occlusion. The results showed that the delivery of CPDC-lipo to the brain ischemic areas could be monitored and quantified by CEST MRI. When administered intra-arterially, CDPC-lipo clearly demonstrated a detectable CEST MRI contrast at 2 ppm. CEST MRI revealed that liposomes preferentially accumulated in the areas of ischemia with a disrupted blood-brain-barrier. We furthermore used CEST MRI to detect the improvement in drug delivery using CDPC-lipo targeted against vascular cell adhesion molecule (VCAM)-1 in the same animal model. The MRI findings were validated using fluorescence microscopy. Hence, liposomal citicoline represents a prototype theranostic system, where the therapeutic agent can be detected directly by CEST MRI in a label-free fashion.


Subject(s)
Cytidine Diphosphate Choline/administration & dosage , Drug Carriers/administration & dosage , Liposomes/administration & dosage , Magnetic Resonance Imaging/methods , Nootropic Agents/administration & dosage , Stroke/diagnostic imaging , Stroke/drug therapy , Animals , Brain/diagnostic imaging , Disease Models, Animal , Injections, Intra-Articular , Rats
10.
Proc Natl Acad Sci U S A ; 111(50): 17977-82, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25404319

ABSTRACT

Melanopsin (opsin4; Opn4), a non-image-forming opsin, has been linked to a number of behavioral responses to light, including circadian photo-entrainment, light suppression of activity in nocturnal animals, and alertness in diurnal animals. We report a physiological role for Opn4 in regulating blood vessel function, particularly in the context of photorelaxation. Using PCR, we demonstrate that Opn4 (a classic G protein-coupled receptor) is expressed in blood vessels. Force-tension myography demonstrates that vessels from Opn4(-/-) mice fail to display photorelaxation, which is also inhibited by an Opn4-specific small-molecule inhibitor. The vasorelaxation is wavelength-specific, with a maximal response at ∼430-460 nm. Photorelaxation does not involve endothelial-, nitric oxide-, carbon monoxide-, or cytochrome p450-derived vasoactive prostanoid signaling but is associated with vascular hyperpolarization, as shown by intracellular membrane potential measurements. Signaling is both soluble guanylyl cyclase- and phosphodiesterase 6-dependent but protein kinase G-independent. ß-Adrenergic receptor kinase 1 (ßARK 1 or GRK2) mediates desensitization of photorelaxation, which is greatly reduced by GRK2 inhibitors. Blue light (455 nM) regulates tail artery vasoreactivity ex vivo and tail blood blood flow in vivo, supporting a potential physiological role for this signaling system. This endogenous opsin-mediated, light-activated molecular switch for vasorelaxation might be harnessed for therapy in diseases in which altered vasoreactivity is a significant pathophysiologic contributor.


Subject(s)
Blood Vessels/physiology , Light , Rod Opsins/metabolism , Signal Transduction/physiology , Vasodilation/physiology , Animals , Blood Vessels/metabolism , Blotting, Western , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Laser-Doppler Flowmetry , Mice , Myography , Reverse Transcriptase Polymerase Chain Reaction , Vasodilation/radiation effects
11.
J Appl Physiol (1985) ; 113(11): 1709-17, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23042910

ABSTRACT

Exchange transfusion of large volumes of hemoglobin (Hb)-based oxygen carriers can protect the brain from middle cerebral artery occlusion (MCAO). Hb in the carboxy state (COHb) may provide protection at relatively low volumes by enhancing vasodilation. We determined whether transfusion of rats with 10 ml/kg PEGylated COHb [polyethylene glycol (PEG)-COHb] at 20 min of 2-h MCAO was more effective in reducing infarct volume compared with non-carbon monoxide (CO) PEG-Hb. After PEG-COHb transfusion, whole blood and plasma COHb was <3%, indicating rapid release of CO. PEG-COHb transfusion significantly reduced infarct volume (15 ± 5% of hemisphere; mean ± SE) compared with that in the control group (35 ± 6%), but non-CO PEG-Hb did not (24 ± 5%). Chemically dissimilar COHb polymers were also effective. Induction of MCAO initially produced 34 ± 2% dilation of pial arterioles in the border region that subsided to 10 ± 1% at 2 h. Transfusion of PEG-COHb at 20 min of MCAO maintained pial arterioles in a dilated state (40 ± 5%) at 2 h, whereas transfusion of non-CO PEG-Hb had an intermediate effect (22 ± 3%). When transfusion of PEG-COHb was delayed by 90 min, laser-Doppler flow in the border region increased from 57 ± 9 to 82 ± 13% of preischemic baseline. These data demonstrate that PEG-COHb is more effective than non-CO PEG-Hb at reducing infarct volume, sustaining cerebral vasodilation, and improving collateral perfusion in a model of transient focal cerebral ischemia when given at a relatively low dose (plasma Hb concentration < 1 g/dl). Use of acellular Hb as a CO donor that is rapidly converted to an oxygen carrier in vivo may permit potent protection at low transfusion volumes.


Subject(s)
Blood Substitutes/pharmacology , Brain/blood supply , Brain/drug effects , Carboxyhemoglobin/pharmacology , Hemoglobins/pharmacology , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/prevention & control , Neuroprotective Agents/pharmacology , Polyethylene Glycols/pharmacology , Animals , Blood Substitutes/administration & dosage , Blood Substitutes/metabolism , Brain/metabolism , Brain/pathology , Brain/physiopathology , Carbon Dioxide/blood , Carboxyhemoglobin/administration & dosage , Carboxyhemoglobin/analogs & derivatives , Carboxyhemoglobin/metabolism , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Hemoglobins/administration & dosage , Hemoglobins/metabolism , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Infusions, Intravenous , Ischemic Attack, Transient/blood , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/metabolism , Rats , Rats, Wistar , Time Factors , Vasodilation/drug effects
12.
Am J Physiol Regul Integr Comp Physiol ; 296(5): R1412-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19261918

ABSTRACT

Partial exchange transfusion with a cell-free hemoglobin (Hb) polymer during transient middle cerebral artery occlusion (MCAO) reduces infarct volume but fails to increase blood flow, as might be expected with the induced decrease in hematocrit. In ischemic brain, endothelin antagonists are known to produce vasodilation. In nonischemic brain, pial arterioles constrict after Hb exchange transfusion, and the constriction is blocked by an inhibitor of 20-HETE synthesis. We tested the hypothesis that a 20-HETE synthesis inhibitor and an endothelin A receptor antagonist increase pial arteriolar dilation after Hb exchange transfusion during MCAO. Pial arteriolar diameter was measured in the ischemic border region of the distal MCA border region through closed cranial windows in anesthetized rats subjected to the filament model of MCAO. During 2 h of MCAO, pial arteriolar dilation gradually subsided from 37 +/- 3 to 7 +/- 5% (+/-SE). Compared with residual dilation at 2 h of MCAO with vehicle superfusion (14 +/- 3%), loss of dilation was not prevented by superfusion of a 20-HETE synthesis inhibitor (21 +/- 5%), partial Hb exchange transfusion (7 +/- 5%) that decreased hematocrit to 23%, or a combination of the two (5 +/- 5%). However, loss of dilation was prevented by superfusion of an endothelin A receptor antagonist with (35 +/- 4%) or without (32 +/- 5%) Hb transfusion. Pial artery constriction during reperfusion was attenuated by HET0016 alone and by BQ610 with or without Hb transfusion. Systemic administration of the endothelin antagonist during prolonged MCAO increased blood flow in the border region. Thus loss of pial arteriolar dilation in the ischemic border region during prolonged MCAO depends on endothelin A receptor activation, and this effect was independent of the presence of cell-free Hb polymers in the plasma. In contrast to previous work in nonischemic brain, inhibition of oxygen-dependent 20-HETE synthesis does not significantly influence the pial arteriolar response to polymeric Hb exchange transfusion during focal ischemia.


Subject(s)
Arterioles/physiology , Brain Ischemia/physiopathology , Endothelins/physiology , Hemoglobins/pharmacology , Hydroxyeicosatetraenoic Acids/physiology , Pia Mater/blood supply , Vasodilation/physiology , Amidines/pharmacology , Animals , Arterioles/drug effects , Blood Substitutes/administration & dosage , Blood Substitutes/pharmacology , Blood Transfusion , Endothelin A Receptor Antagonists , Endothelins/antagonists & inhibitors , Hemoglobins/administration & dosage , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Male , Models, Animal , Oligopeptides/pharmacology , Polymers , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Vasodilation/drug effects
13.
Am J Physiol Regul Integr Comp Physiol ; 291(3): R728-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16601261

ABSTRACT

Nitric oxide (NO) has been implicated in mediation of cerebral vasodilation during neuronal activation and, specifically, in pharmacological activation of N-methyl-d-aspartate (NMDA) and kainate receptors. Possible mediators of cerebral vasodilation to alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) have not been well studied in mature brain, although heme oxygenase (HO) activity has been implicated in newborn pigs. In anesthetized rats, 5 min of topical superfusion of 30 and 100 microM AMPA on the cortical surface through a closed cranial window resulted in increases in pial arteriolar diameter. The dilatory response to AMPA was not inhibited by superfusion of an NO synthase inhibitor, a cyclooxygenase-2 inhibitor, or a cytochrome P-450 epoxygenase inhibitor, all of which have been shown to inhibit the cortical blood flow response to sensory activation. However, the 48 +/- 13% dilation to 100 microM AMPA was attenuated 56-71% by superfusion of the adenosine A(2A) receptor antagonist ZM-241385, the A(2B) receptor antagonist alloxazine, and the HO inhibitor chromium mesoporphyrin. Combination of the latter three inhibitors did not attenuate the dilator response more than the individual inhibitors, whereas an AMPA receptor antagonist fully blocked the vasodilation to AMPA. These results indicate that cortical pial arteriolar dilation to AMPA does not require activation of NO synthase, cyclooxygenase-2, or cytochrome P-450 epoxygenase but does depend on activation of adenosine A(2A) and A(2B) receptors. In addition, CO derived from HO appears to play a role in the vascular response to AMPA receptor activation in mature brain by a mechanism that is not additive with that of adenosine receptor activation.


Subject(s)
Arterioles/physiology , Heme Oxygenase (Decyclizing)/metabolism , Pia Mater/blood supply , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Vasodilation/physiology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Animals , Arterioles/drug effects , Male , Rats , Rats, Wistar , Time Factors , Vasodilation/drug effects
14.
Am J Physiol Heart Circ Physiol ; 290(3): H1027-37, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16214847

ABSTRACT

Polymers of cell-free hemoglobin have been designed for clinical use as oxygen carriers, but limited information is available regarding their effects on vascular regulation. We tested the hypothesis that the contribution of heme oxygenase (HO) to acetylcholine-evoked dilation of pial arterioles is upregulated 2 days after polymeric hemoglobin transfusion. Dilator responses to acetylcholine measured by intravital microscopy in anesthetized cats were blocked by superfusion of the HO inhibitor tin protoporphyrin-IX (SnPPIX) in a group that had undergone exchange transfusion with hemoglobin 2 days earlier but not in surgical sham and albumin-transfused groups. However, immunoblots from cortical brain homogenates did not reveal changes in expression of the inducible isoform HO1 or the constitutive isoform HO2 in the hemoglobin-transfused group. To test whether the inhibitory effect of SnPPIX was present acutely after hemoglobin transfusion, responses were measured within an hour of completion of the exchange transfusion. In control and albumin-transfused groups, acetylcholine responses were unaffected by SnPPIX but were blocked by addition of the nitric oxide synthase inhibitor N(omega)-nitro-l-arginine (l-NNA) to the superfusate. In hemoglobin-transfused groups, the acetylcholine response was blocked by either SnPPIX or l-NNA alone. The effect of another HO inhibitor, chromium mesoporphyrin (CrMP), was tested on ADP, another endothelial-dependent dilator, in anesthetized rats. Pial arteriolar dilation to ADP was unaffected by CrMP in controls but was attenuated 62% by CrMP in rats transfused with hemoglobin. It is concluded that 1) polymeric hemoglobin transfusion acutely upregulates the contribution of HO to acetylcholine-induced dilation of pial arterioles in cats, 2) this upregulation persists 2 days after transfusion when 95% of the hemoglobin is cleared from the circulation, and 3) this acute upregulation of HO signaling is ubiquitous in that similar effects were observed with a different endothelial-dependent agonist (i.e., ADP) in a another species (rat).


Subject(s)
Acetylcholine/metabolism , Adenosine Diphosphate/metabolism , Arterioles/metabolism , Blood Substitutes/administration & dosage , Heme Oxygenase (Decyclizing)/metabolism , Hemoglobins/administration & dosage , Pia Mater/blood supply , Pia Mater/metabolism , Vasodilation/physiology , Animals , Arterioles/drug effects , Blood Transfusion , Cats , Male , Pia Mater/drug effects , Vasodilation/drug effects
15.
Am J Physiol Heart Circ Physiol ; 282(5): H1778-86, 2002 May.
Article in English | MEDLINE | ID: mdl-11959643

ABSTRACT

We investigated whether oxygen radicals generated during ischemia-reperfusion trigger postischemic inflammation in the heart. Closed-chest dogs underwent 90-min coronary artery occlusion, followed by 1- or 3-h reperfusion: 10 dogs received the cell-permeant oxygen radical scavenger N-(2-mercaptopropionyl)-glycine (MPG; 8 mg x kg(-1) x h(-1) intracoronary) beginning 5 min before reperfusion, and 9 dogs received vehicle. Blood flow (microspheres), intercellular adhesion molecule (ICAM)-1 protein expression (immunohistochemistry), ICAM-1 gene activation (Northern blotting), nuclear DNA binding activity of nuclear factor (NF)-kappaB and AP-1 (electrophoretic mobility shift assays), and neutrophil (PMN) accumulation (myeloperoxidase activity) were assessed in myocardial tissue samples. ICAM-1 protein expression was high in vascular endothelium after ischemia-reperfusion but was markedly reduced by MPG. MPG treatment also markedly decreased expression of ICAM-1 mRNA and tissue PMN accumulation. Nuclear DNA binding activities of NF-kappaB and AP-1, increased by ischemia-reperfusion, were both markedly decreased by MPG at 1 h of reperfusion. However, by 3 h, AP-1 activity was only modestly reduced by MPG and NF-kappaB activity was not significantly different from ischemic-reperfused controls. These results suggest that oxygen radicals generated in vivo during reperfusion trigger early activation of NF-kappaB and AP-1, resulting in upregulation of the ICAM-1 gene in vascular endothelium and subsequent tissue accumulation of activated PMNs.


Subject(s)
Glycine/analogs & derivatives , Intercellular Adhesion Molecule-1/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion , NF-kappa B/metabolism , Reactive Oxygen Species/pharmacology , Transcription Factor AP-1/metabolism , Animals , Blood Flow Velocity , Blotting, Northern , Constriction , Coronary Vessels , DNA/metabolism , Dogs , Free Radical Scavengers , Free Radicals , Gene Expression Regulation , Glycine/pharmacology , Hemodynamics , Immunohistochemistry , Intercellular Adhesion Molecule-1/analysis , Intercellular Adhesion Molecule-1/genetics , Myocardial Ischemia/pathology , Neutrophils/pathology , Neutrophils/physiology , Peroxidase/metabolism , RNA, Messenger/analysis , Sulfhydryl Compounds/pharmacology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...