Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 133
1.
Neurochem Int ; 162: 105462, 2023 01.
Article En | MEDLINE | ID: mdl-36509234

Diabetes exacerbates hemorrhagic transformation (HT) after stroke and worsens clinical outcomes. Female patients with diabetes are at a greater risk of stroke and worsened recovery. We have shown that activation of matrix metalloprotease 3 (MMP3) in hyperglycemic settings mediates HT in male rats. In light of our recent findings that diabetic female rats develop greater HT, the current study was designed to test the hypotheses that: 1) cerebral microvascular MMP3 activation contributes to poor functional outcomes and increased hemorrhagic transformations (HT) after ischemic stroke, and 2) MMP3 inhibition can improve functional outcomes in female diabetic rats. Female control and diabetic Wistar rats were subjected to 60 min of middle cerebral artery occlusion (MCAO). One cohort of diabetic animals received a single dose of MMP3 inhibitor (UK356618; 15 mg/kg; iv) or vehicle after reperfusion. Neurobehavioral outcomes, brain infarct size, edema, HT, and MMPs were measured in brain tissue. Diabetic rats had significant neurological deficits on Day 3 after stroke. MMP3 expression and enzyme activity were significantly increased in both micro and macro vessels of diabetic animals. MMP3 inhibition improved functional outcomes and reduced brain edema and HT scores. In conclusion, cerebral endothelial MMP3 activation to vascular injury in female diabetic rats. Our findings identify MMP3 as a potential therapeutic target in diabetic stroke.


Diabetes Mellitus, Experimental , Infarction, Middle Cerebral Artery , Matrix Metalloproteinase 3 , Microvessels , Stroke , Vascular System Injuries , Animals , Female , Male , Rats , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Matrix Metalloproteinase 3/metabolism , Rats, Wistar , Stroke/complications , Vascular System Injuries/enzymology , Vascular System Injuries/etiology , Microvessels/enzymology , Cerebrum/blood supply
2.
Am J Physiol Cell Physiol ; 322(3): C338-C353, 2022 03 01.
Article En | MEDLINE | ID: mdl-35044858

The small conductance calcium-activated potassium channel (KCa2.3) has long been recognized for its role in mediating vasorelaxation through the endothelium-derived hyperpolarization (EDH) response. Histone deacetylases (HDACs) have been implicated as potential modulators of blood pressure and histone deacetylase inhibitors (HDACi) are being explored as therapeutics for hypertension. Herein, we show that HDACi increase KCa2.3 expression when heterologously expressed in HEK cells and endogenously expressed in primary cultures of human umbilical vein endothelial cells (HUVECs) and human intestinal microvascular endothelial cells (HIMECs). When primary endothelial cells were exposed to HDACi, KCa2.3 transcripts, subunits, and functional current are increased. Quantitative RT-PCR (qPCR) demonstrated increased KCa2.3 mRNA following HDACi, confirming transcriptional regulation of KCa2.3 by HDACs. By using pharmacological agents selective for different classes of HDACs, we discriminated between cytoplasmic and epigenetic modulation of KCa2.3. Biochemical analysis revealed an association between the cytoplasmic HDAC6 and KCa2.3 in immunoprecipitation studies. Specifically inhibiting HDAC6 increases expression of KCa2.3. In addition to increasing the expression of KCa2.3, we show that nonspecific inhibition of HDACs causes an increase in the expression of the molecular chaperone Hsp70 in endothelial cells. When Hsp70 is inhibited in the presence of HDACi, the magnitude of the increase in KCa2.3 expression is diminished. Finally, we show a slower rate of endocytosis of KCa2.3 as a result of exposure of primary endothelial cells to HDACi. These data provide the first demonstrated approach to increase KCa2.3 channel number in endothelial cells and may partially account for the mechanism by which HDACi induce vasorelaxation.


Endothelial Cells/drug effects , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Intestines/blood supply , Microvessels/drug effects , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Endocytosis , Endothelial Cells/enzymology , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Histone Deacetylase 6/metabolism , Humans , Membrane Potentials , Microvessels/enzymology , Small-Conductance Calcium-Activated Potassium Channels/genetics , Up-Regulation , Vasodilation
3.
Biosci Rep ; 42(1)2022 01 28.
Article En | MEDLINE | ID: mdl-34750610

BACKGROUND: Acute lung injury (ALI) is a respiratory disease with high morbidity and mortality rates. Currently, there is no effective treatment to complement mechanical ventilation. Exosomes and microRNAs (miRNAs) are promising agents for the management of this disease. METHODS: Exosomes were isolated from mouse bone marrow stromal stem cells (BMSCs). The levels of two miRNAs, miR-542-3P and miR-150, in exosomes were determined using RT-PCR, and miR-150 was selected for further study. ALI model was established in mice using lipopolysaccharides, and then, they were treated with saline, exosomes, miRNA agomirs, or miRNA antagomirs. The concentrations of TNF-α, IL-6, and IL-1ß and the number of neutrophils and macrophages in the bronchoalveolar lavage fluid were measured. The wet/dry weight ratio of the lung tissue was calculated, and tissue pathology and apoptosis were observed using hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. CD34 and VE-cadherin expression was detected using immunofluorescence. Proteins associated with apoptosis and MAPK signaling were detected using Western blotting, and miR-150 expression in lung tissue was evaluated using RT-PCR. RESULTS: We successfully isolated BMSCs and exosomes and showed that the level of miR-150 was significantly higher than that of miR-542-3p. Exosomes and miR-150 reduced inflammation and lung edema while maintaining the integrity of the alveolar structure. They also mitigated microvascular endothelial cell injury by regulating the caspase-3, Bax/Bcl-2, and MAPK signaling. CONCLUSIONS: Exosomal miR-150 attenuates lipopolysaccharide-induced ALI through the MAPK pathway.


Acute Lung Injury/prevention & control , Exosomes/transplantation , Lung/blood supply , Mesenchymal Stem Cell Transplantation , MicroRNAs/metabolism , Microvessels/enzymology , Mitogen-Activated Protein Kinases/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Exosomes/genetics , Exosomes/metabolism , Lipopolysaccharides , Lung/enzymology , Lung/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , Signal Transduction
4.
Open Heart ; 8(2)2021 11.
Article En | MEDLINE | ID: mdl-34819349

BACKGROUND: Myocardial infarction (MI) is associated with mental health disorders, in which neuroinflammation and cerebral microvascular dysfunction may play a role. Previously, we have shown that the proinflammatory factors Nε-(carboxymethyl)lysine (CML) and NADPH oxidase 2 (NOX2) are increased in the human infarcted heart microvasculature. The aim of this study was to analyse the presence of CML and NOX2 in the cerebral microvasculature of patients with MI. METHODS: Brain tissue was obtained at autopsy from 24 patients with MI and nine control patients. According to their infarct age, patients with MI were divided into three groups: 3-6 hours old (phase I), 6 hours-5 days old (phase II) and 5-14 days old (phase III). CML and NOX2 in the microvasculature were studied through immunohistochemical analysis. RESULTS: We observed a 2.5-fold increase in cerebral microvascular CML in patients with phase II and phase III MI (phase II: 21.39±7.91, p=0.004; phase III: 24.21±10.37, p=0.0007) compared with non-MI controls (8.55±2.98). NOX2 was increased in microvessels in patients with phase II MI (p=0.002) and phase III MI (p=0.04) compared with controls. No correlation was found between CML and NOX2 (r=0.58, p=0.13). CONCLUSIONS: MI coincides with an increased presence of CML and NOX2 in the brain microvasculature. These data point to proinflammatory alterations in the brain microvasculature that may underlie MI-associated mental health disorders.


Cerebral Arteries/enzymology , Lysine/analogs & derivatives , Microvessels/enzymology , Myocardial Infarction/enzymology , NADPH Oxidase 2/biosynthesis , Neuroinflammatory Diseases/enzymology , Aged , Biomarkers/metabolism , Cerebral Arteries/pathology , Female , Humans , Immunohistochemistry , Lysine/biosynthesis , Male , Microvessels/pathology , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/pathology , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/pathology
5.
Am J Physiol Cell Physiol ; 321(6): C964-C977, 2021 12 01.
Article En | MEDLINE | ID: mdl-34586897

Pulmonary microvascular endothelial cells (PMVECs) uniquely express an α1G-subtype of voltage-gated T-type Ca2+ channel. We have previously revealed that the α1G channel functions as a background Ca2+ entry pathway that is critical for the cell proliferation, migration, and angiogenic potential of PMVECs, a novel function attributed to the coupling between α1G-mediated Ca2+ entry and constitutive Akt phosphorylation and activation. Despite this significance, mechanism(s) that link the α1G-mediated Ca2+ entry to Akt phosphorylation remain incompletely understood. In this study, we demonstrate that Ca2+/calmodulin-dependent protein kinase (CaMK) 4 serves as a downstream effector of the α1G-mediated Ca2+ entry to promote the angiogenic potential of PMVECs. Notably, CaMK2 and CaMK4 are both expressed in PMVECs. Pharmacological blockade or genetic knockdown of the α1G channel led to a significant reduction in the phosphorylation level of CaMK4 but not the phosphorylation level of CaMK2. Pharmacological inhibition as well as genetic knockdown of CaMK4 significantly decreased cell proliferation, migration, and network formation capacity in PMVECs. However, CaMK4 inhibition or knockdown did not alter Akt phosphorylation status in PMVECs, indicating that α1G/Ca2+/CaMK4 is independent of the α1G/Ca2+/Akt pathway in sustaining the cells' angiogenic potential. Altogether, these findings suggest a novel α1G-CaMK4 signaling complex that regulates the Ca2+-dominated angiogenic potential in PMVECs.


Calcium Channels, T-Type/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Calcium/metabolism , Endothelial Cells/enzymology , Lung/blood supply , Microvessels/enzymology , Neovascularization, Physiologic , Angiogenesis Inhibitors/pharmacology , Animals , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/drug effects , Male , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley
6.
Am J Physiol Heart Circ Physiol ; 321(5): H985-H1003, 2021 11 01.
Article En | MEDLINE | ID: mdl-34559580

Ventilation with gases containing enhanced fractions of oxygen is the cornerstone of therapy for patients with hypoxia and acute respiratory distress syndrome. Yet, hyperoxia treatment increases free reactive oxygen species (ROS)-induced lung injury, which is reported to disrupt autophagy/mitophagy. Altered extranuclear activity of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), plays a protective role in ROS injury and autophagy in the systemic and coronary endothelium. We investigated interactions between autophagy/mitophagy and TERT that contribute to mitochondrial dysfunction and pulmonary injury in cultured rat lung microvascular endothelial cells (RLMVECs) exposed in vitro, and rat lungs exposed in vivo to hyperoxia for 48 h. Hyperoxia-induced mitochondrial damage in rat lungs [TOMM20, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], which was paralleled by increased markers of inflammation [myeloperoxidase (MPO), IL-1ß, TLR9], impaired autophagy signaling (Beclin-1, LC3B-II/1, and p62), and decreased the expression of TERT. Mitochondrial-specific autophagy (mitophagy) was not altered, as hyperoxia increased expression of Pink1 but not Parkin. Hyperoxia-induced mitochondrial damage (TOMM20) was more pronounced in rats that lack the catalytic subunit of TERT and resulted in a reduction in cellular proliferation rather than cell death in RLMVECs. Activation of TERT or autophagy individually offset mitochondrial damage (MTT). Combined activation/inhibition failed to alleviate hyperoxic-induced mitochondrial damage in vitro, whereas activation of autophagy in vivo decreased mitochondrial damage (MTT) in both wild type (WT) and rats lacking TERT. Functionally, activation of either TERT or autophagy preserved transendothelial membrane resistance. Altogether, these observations show that activation of autophagy/mitophagy and/or TERT mitigate loss of mitochondrial function and barrier integrity in hyperoxia.NEW & NOTEWORTHY In cultured pulmonary artery endothelial cells and in lungs exposed in vivo to hyperoxia, autophagy is activated, but clearance of autophagosomes is impaired in a manner that suggests cross talk between TERT and autophagy. Stimulation of autophagy prevents hyperoxia-induced decreases in mitochondrial metabolism and sustains monolayer resistance. Hyperoxia increases mitochondrial outer membrane (TOMM20) protein, decreases mitochondrial function, and reduces cellular proliferation without increasing cell death.


Endothelial Cells/enzymology , Hyperoxia/complications , Lung Injury/enzymology , Lung/blood supply , Microvessels/enzymology , Mitochondria/enzymology , Mitophagy , Telomerase/metabolism , Animals , Autophagy-Related Proteins/metabolism , Capillary Permeability , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Female , Gene Knockout Techniques , Inflammation Mediators/metabolism , Lung Injury/etiology , Lung Injury/genetics , Lung Injury/pathology , Male , Membrane Transport Proteins/metabolism , Microvessels/pathology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Precursor Protein Import Complex Proteins , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, Cell Surface/metabolism , Telomerase/deficiency , Telomerase/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
7.
Microvasc Res ; 138: 104227, 2021 11.
Article En | MEDLINE | ID: mdl-34324883

This study examined the effects of vitamin D deficiency on vascular function and tissue oxidative status in the microcirculation; and whether or not these effects can be ameliorated with calcitriol, the active vitamin D metabolite. Three groups (n = 10 each) of male Sprague Dawley rats were fed for 10 weeks with control diet (CR), vitamin D-deficient diet without (DR), or with oral calcitriol supplementation (0.15 µg/kg) for the last four weeks (DSR). After 10 weeks, rats were sacrificed; mesenteric arterial rings were studied using wire myograph. Oxidative stress biomarkers malondialdehyde (MDA) levels and superoxide dismutase (SOD) activity were measured in the mesenteric arterial tissue. Vascular protein expression of endothelial nitric oxide synthase (eNOS) was determined by Western blotting. Acetylcholine-induced endothelium-dependent relaxation of DR was lower than CR. eNOS expression and SOD activity were lower in mesenteric arterial tissue of DR compared to CR. Calcitriol supplementation to DSR did not ameliorate the above parameters; in fact, augmented endothelium-dependent contraction was observed. Serum calcium was higher in DSR compared to CR and DR. In conclusion, vitamin D deficiency impaired microvascular vasodilation, associated with eNOS downregulation and reduced antioxidant activity. Calcitriol supplementation to vitamin D-deficient rats at the dosage used augmented endothelium-dependent contraction, possibly due to hypercalcaemia.


Antioxidants/metabolism , Endothelium, Vascular/enzymology , Microcirculation , Microvessels/enzymology , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Vasodilation , Vitamin D Deficiency/enzymology , Animals , Calcitriol/pharmacology , Calcium/blood , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Male , Malondialdehyde/metabolism , Microcirculation/drug effects , Microvessels/drug effects , Microvessels/physiopathology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction , Superoxide Dismutase/metabolism , Vasodilation/drug effects , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/physiopathology , Vitamins/pharmacology
8.
J Thorac Cardiovasc Surg ; 162(2): e143-e156, 2021 08.
Article En | MEDLINE | ID: mdl-32414595

BACKGROUND: c-Jun N-terminal kinase 1 (JNK1) and JNK2 regulate distinct pathological processes in lung diseases. Here we discriminated the respective roles of these kinases in lung transplantation-induced ischemia-reperfusion injury (IRI). METHODS: Rat pulmonary microvascular endothelial cells were transfected with JNK1 small-interfering RNA (siRNA) and JNK2 siRNA and then subjected to in vitro IRI. For the isoform confirmed to aggravate IRI, the delivery of short-hairpin RNA (shRNA) plasmid was performed by intratracheal administration 48 hours before transplantation into donor rats. After a 3-hour reperfusion, the samples were collected. RESULTS: JNK1 siRNA decreased but JNK2 siRNA increased JNK phosphorylation and activity, phosphorylated and total c-Jun, and activator protein-1 activity. Although JNK1 siRNA decreased apoptosis and the levels of malondialdehyde, interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF-α), it increased the levels of superoxide dismutase, S-phase percentage, and cyclin D1; JNK2 siRNA had a converse effect. JNK1 siRNA decreased the level of lactate dehydrogenase and increased the levels of VE-cadherin, nitric oxide, phosphorylated nitric oxide synthase, and cell viability; JNK2 si RNA had a converse effect. Compared with the control group, the JNK1 shRNA group exhibited a higher lung oxygenation index and lower lung apoptosis index, injury score, wet weight:dry weight ratio, and levels of IL-1, IL-6, and TNF-α. CONCLUSIONS: JNK1 aggravated, but JNK2 alleviated, IRI through differential regulation of the JNK1 pathway in in vitro ischemia-reperfusion. JNK1 silence attenuated lung graft dysfunction by inhibiting inflammation and apoptosis. These findings provide a theoretical basis for devising therapeutic strategies against IRI after lung transplantation.


Endothelial Cells/enzymology , Lung/blood supply , Microvessels/enzymology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Reperfusion Injury/enzymology , Transcription Factor AP-1/metabolism , Animals , Apoptosis , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Inflammation Mediators/metabolism , Isoenzymes , Lung Transplantation/adverse effects , Microvessels/pathology , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Phosphorylation , Rats , Reperfusion Injury/etiology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
9.
Microvasc Res ; 133: 104078, 2021 01.
Article En | MEDLINE | ID: mdl-32980388

The present study assessed the impact of impaired tetrahydrobiopterin (BH4) production on vasoreactivity from conduit and small arteries along the vascular tree as seen during aging. For this purpose, the mutant hyperphenylalaninemic mouse (hph-1) was used. This model is reported to be deficient in GTP cyclohydrolase I, a rate limiting enzyme in BH4 biosynthesis. BH4 is a key regulator of vascular homeostasis by regulating the nitric oxide synthase 3 (NOS3) activity. In GTP-CH deficient mice, the aortic BH4 levels were decreased, by -77% in 12 week-middle-aged mice (young) and by -83% in 35-45 week-middle-aged mice (middle-aged). In young hph-1, the mesenteric artery ability to respond to flow was slightly reduced by 9%. Aging induced huge modification in many vascular functions. In middle-aged hph-1, we observed a decrease in aortic cGMP levels, biomarker of NO availability (-46%), in flow-mediated vasodilation of mesenteric artery (-31%), in coronary hyperemia response measured in isolated heart following transient ischemia (-27%) and in cutaneous microcirculation dilation in response to acetylcholine assessed in vivo by laser-doppler technic (-69%). In parallel, the endothelium-dependent relaxation in response to acetylcholine in conduit blood vessel, measured on isolated aorta rings, was unchanged in hph-1 mice whatever the age. Our findings demonstrate that in middle-aged GTP-CH depleted mice, the reduction of BH4 was characterized by an alteration of microcirculation dilatory properties observed in various parts of the vascular tree. Large conduit blood vessels vasoreactivity, ie aorta, was unaltered even in middle-aged mice emphasizing the main BH4-deletion impact on the microcirculation.


GTP Cyclohydrolase/deficiency , Microcirculation , Microvessels/enzymology , Phenylketonurias/enzymology , Skin/blood supply , Vasodilation , Age Factors , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Biopterins/analogs & derivatives , Biopterins/metabolism , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Disease Models, Animal , GTP Cyclohydrolase/genetics , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microvessels/physiopathology , Phenylketonurias/genetics , Phenylketonurias/physiopathology
10.
Radiat Res ; 194(6): 625-635, 2020 12 01.
Article En | MEDLINE | ID: mdl-33348373

Persistent vasculature abnormalities contribute to an altered CNS microenvironment that further compromises the integrity of the blood-brain barrier and exposes the brain to a host of neurotoxic conditions. Standard radiation therapy at conventional (CONV) dose rate elicits short-term damage to the blood-brain barrier by disrupting supportive cells, vasculature volume and tight junction proteins. While current clinical applications of cranial radiotherapy use dose fractionation to reduce normal tissue damage, these treatments still cause significant complications. While dose escalation enhances treatment of radiation-resistant tumors, methods to subvert normal tissue damage are clearly needed. In this regard, we have recently developed a new modality of irradiation based on the use of ultra-high-dose-rate FLASH that does not induce the classical pathogenic patterns caused by CONV irradiation. In previous work, we optimized the physical parameters required to minimize normal brain toxicity (i.e., FLASH, instantaneous intra-pulse dose rate, 6.9 · 106 Gy/s, at a mean dose rate of 2,500 Gy/s), which we then used in the current study to determine the effect of FLASH on the integrity of the vasculature and the blood-brain barrier. Both early (24 h, one week) and late (one month) timepoints postirradiation were investigated using C57Bl/6J female mice exposed to whole-brain irradiation delivered in single doses of 25 Gy and 10 Gy, respectively, using CONV (0.09 Gy/s) or FLASH (>106 Gy/s). While the majority of changes found one day postirradiation were minimal, FLASH was found to reduce levels of apoptosis in the neurogenic regions of the brain at this time. At one week and one month postirradiation, CONV was found to induce vascular dilation, a well described sign of vascular alteration, while FLASH minimized these effects. These results were positively correlated with and temporally coincident to changes in the immunostaining of the vasodilator eNOS colocalized to the vasculature, suggestive of possible dysregulation in blood flow at these latter times. Overall expression of the tight junction proteins, occludin and claudin-5, which was significantly reduced after CONV irradiation, remained unchanged in the FLASH-irradiated brains at one and four weeks postirradiation. Our data further confirm that, compared to isodoses of CONV irradiation known to elicit detrimental effects, FLASH does not damage the normal vasculature. These data now provide the first evidence that FLASH preserves microvasculature integrity in the brain, which may prove beneficial to cognition while allowing for better tumor control in the clinic.


Enzyme Induction/radiation effects , Nitric Oxide Synthase Type III/biosynthesis , Radiotherapy/methods , Tight Junctions/radiation effects , Vasodilation/radiation effects , Animals , Apoptosis/radiation effects , Female , Mice , Mice, Inbred C57BL , Microvessels/enzymology , Microvessels/pathology , Microvessels/radiation effects
11.
Basic Res Cardiol ; 115(6): 64, 2020 10 14.
Article En | MEDLINE | ID: mdl-33057972

By cleaving sphingomyelin into ceramide, which is an essential component of plasma membrane microdomains, acid sphingomyelinase (Asm) pivotally controls cell signaling. To define how the activation of the Asm/ceramide pathway, which occurs within seconds to minutes upon stress stimuli, influences brain ischemia/reperfusion (I/R) injury, we exposed male and female wildtype mice carrying both alleles of Asm's gene sphingomyelinase phosphodiesterase-1 (Smpd1+/+), heterozygously Asm-deficient mice (Smpd1+/-) and homozygously Asm-deficient mice (Smpd1-/-) of different age (8, 12 or 16 weeks) to 30, 60 or 90 min intraluminal middle cerebral artery occlusion (MCAO). For studying the contribution of brain-invading polymorphonuclear neutrophils (PMN) to I/R injury, PMNs were depleted by delivery of a PMN-specific Ly6G antibody. In male and female mice exposed to 30 min, but not 60 or 90 min MCAO, homozygous Smpd1-/- consistently increased I/R injury, blood-brain barrier permeability and brain leukocyte and PMN infiltration, whereas heterozygous Smpd1+/- reduced I/R injury. Increased abundance of the intercellular leukocyte adhesion molecule ICAM-1 was noted on cerebral microvessels of Smpd1-/- mice. PMN depletion by anti-Ly6G delivery prevented the exacerbation of I/R injury in Smpd1-/- compared with wildtype mice and reduced brain leukocyte infiltrates. Our results show that Asm tempers leukocyte entry into the reperfused ischemic brain, thereby attenuating I/R injury.


Brain/blood supply , Infarction, Middle Cerebral Artery/enzymology , Microvessels/enzymology , Neutrophil Infiltration , Neutrophils/enzymology , Reperfusion Injury/enzymology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Disease Models, Animal , Female , Heterozygote , Homozygote , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Sphingomyelin Phosphodiesterase/genetics , Time Factors
12.
Microcirculation ; 27(6): e12624, 2020 08.
Article En | MEDLINE | ID: mdl-32352607

OBJECTIVE: Inhibition of adenosine kinase (ADK), via augmenting endogenous adenosine levels exerts cardiovascular protection. We tested the hypothesis that ADK inhibition improves microvascular dilator and left ventricle (LV) contractile function under metabolic or hemodynamic stress. METHODS AND RESULTS: In Obese diabetic Zucker fatty/spontaneously hypertensive heart failure F1 hybrid rats, treatment with the selective ADK inhibitor, ABT-702 (1.5 mg/kg, intraperitoneal injections for 8-week) restored acetylcholine-, sodium nitroprusside-, and adenosine-induced dilations in isolated coronary arterioles, an effect that was accompanied by normalized end-diastolic pressure (in mm Hg, Lean: 3.4 ± 0.6, Obese: 17.6 ± 4.2, Obese + ABT: 6.6 ± 1.4) and LV relaxation constant, Tau (in ms, Lean: 6.9 ± 1.5, Obese: 13.9 ± 1.7, Obese + ABT: 6.0 ± 1.1). Mice with vascular endothelium selective ADK deletion (ADKVEC KO) exhibited an enhanced dilation to acetylcholine in isolated gracilis muscle (lgEC50 WT: -8.2 ± 0.1, ADKVEC KO: -8.8 ± 0.1, P < .05) and mesenteric arterioles (lgEC50 WT: -7.4 ± 0.2, ADKVEC KO: -8.1 ± 1.2, P < .05) when compared to wild-type (WT) mice, whereas relaxation of the femoral artery and aorta (lgEC50 WT: -7.03 ± 0.6, ADKVEC KO: -7.05 ± 0.8) was similar in the two groups. Wild-type mice progressively developed LV systolic and diastolic dysfunction when they underwent transverse aortic constriction surgery, whereas ADKVEC -KO mice displayed a lesser degree in decline of LV function. CONCLUSIONS: Our results indicate that ADK inhibition selectively enhances microvascular vasodilator function, whereby it improves LV perfusion and LV contractile function under metabolic and hemodynamic stress.


Adenosine Kinase/antagonists & inhibitors , Microvessels/enzymology , Morpholines/pharmacology , Pyrimidines/pharmacology , Vasodilation/drug effects , Ventricular Dysfunction, Left/enzymology , Adenosine Kinase/genetics , Adenosine Kinase/metabolism , Animals , Diastole/drug effects , Diastole/genetics , Male , Mice , Mice, Knockout , Rats , Rats, Zucker , Vasodilation/genetics , Ventricular Dysfunction, Left/genetics
13.
Microvasc Res ; 131: 104012, 2020 09.
Article En | MEDLINE | ID: mdl-32428522

Recent evidences have shown that reactive oxygen species (ROS) are involved in regulating angiogenesis and preventing tissue injury. However, the precise molecular mechanisms behind ROS-induced angiogenesis are still unknown. The aim of the present study was to investigate the effects of ROS-induced angiogenesis in rat brain microvessel endothelial cells (rBMECs) and identify involving the signal pathways. For initial experiments, the rBMECs were incubated with different concentrations of hydrogen peroxide (H2O2). For the second experiments, the rBMECs were respectively treated with ROS scavenger dimethylthiourea (DMTU), NADPH oxidase (Nox) inhibitor apocynin, small interfering RNAs-mediated knock down Nox2 or Nox4, or pretreated with c-Jun N-terminal kinase (JNK) inhibitor SP600125. The cell proliferation, migration, tube formation, and the expressions of several important neuroangiogenic factors including vascular endothelial growth factor (VEGF), brain derived neurotrophic factor (BDNF), matrix metalloproteinase (MMP) -9 and phos-JNK were measured. Low level of H2O2 significantly promoted endothelial cell (EC) proliferation, migration and tube formation and upregulated levels of VEGF, BDNF, MMP-9 and phos-JNK. DMTU and apocynin significantly inhibited endothelial angiogenesis and downregulated these protein levels. As expected, knockdown of Nox2 or Nox4 expression blocked endothelial angiogenesis and downregulated the expressions of pro-neuroangiogenic factors. Furthermore, H2O2-induced endothelial angiogenesis and high expressions of pro-neuroangiogenic factors were decreased by SP600125. In conclusion, Nox-derived ROS were required for endothelial angiogenesis. Low level of ROS may activate JNK signaling pathway and upregulate pro-neuroangiogenic factors, ultimately mediating endothelial angiogenesis.


Cerebral Cortex/blood supply , Endothelial Cells/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Microvessels/enzymology , NADP/metabolism , Neovascularization, Physiologic , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Microvessels/drug effects , NADPH Oxidase 2/antagonists & inhibitors , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Neovascularization, Physiologic/drug effects , Oxidants/pharmacology , Phosphorylation , Rats , Signal Transduction
14.
Vascul Pharmacol ; 128-129: 106677, 2020.
Article En | MEDLINE | ID: mdl-32240815

Acute Respiratory Distress Syndrome (ARDS) is a devastating disease process that involves dysregulated inflammation and decreased alveolar-capillary barrier function. Despite increased understanding of the pathophysiology, no effective targeted therapies exist to treat ARDS. Recent preclinical studies suggest that the multi-tyrosine kinase inhibitor, imatinib, which targets the Abl kinases c-Abl and Arg, has the potential to restore endothelial dysfunction caused by inflammatory agonists. Prior work demonstrates that imatinib attenuates LPS (lipopolysaccharide)-induced vascular leak and inflammation; however, the mechanisms underlying these effects remain incompletely understood. In the current study, we demonstrate that imatinib inhibits LPS-induced increase in the phosphorylation of CrkL, a specific substrate of Abl kinases, in human pulmonary endothelial cells. Specific silencing of Arg, and not c-Abl, attenuated LPS-induced pulmonary vascular permeability as measured by electrical cellular impedance sensing (ECIS) and gap formation assays. In addition, direct activation of Abl family kinases with the small molecule activator DPH resulted in endothelial barrier disruption that was attenuated by Arg siRNA. In complementary studies to characterize the mechanisms by which Arg mediates endothelial barrier function, Arg silencing was found to inhibit LPS-induced disruption of adherens junctions and phosphorylation of myosin light chains (MLC). Overall, these results characterize the mechanisms by which imatinib protects against LPS-induced endothelial barrier disruption and suggest that Arg inhibition may represent a novel strategy to enhance endothelial barrier function.


Capillary Permeability/drug effects , Lipopolysaccharides/pharmacology , Microvessels/drug effects , Protein-Tyrosine Kinases/metabolism , Pulmonary Artery/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/drug effects , Adherens Junctions/enzymology , Adherens Junctions/pathology , Cells, Cultured , Electric Impedance , Humans , Microvessels/enzymology , Microvessels/pathology , Myosin Light Chains/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Signal Transduction
15.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R390-R398, 2020 02 01.
Article En | MEDLINE | ID: mdl-31913684

Local skin heating to 42°C causes cutaneous thermal hyperemia largely via nitric oxide (NO) synthase (NOS)-related mechanisms. We assessed the hypothesis that ATP-sensitive K+ (KATP) channels interact with NOS to mediate cutaneous thermal hyperemia. In 13 young adults (6 women, 7 men), cutaneous vascular conductance (CVC) was measured at four intradermal microdialysis sites that were continuously perfused with 1) lactated Ringer solution (control), 2) 5 mM glibenclamide (KATP channel blocker), 3) 20 mM NG-nitro-l-arginine methyl ester (NOS inhibitor), or 4) a combination of KATP channel blocker and NOS inhibitor. Local skin heating to 42°C was administered at all four treatment sites to elicit cutaneous thermal hyperemia. Thirty minutes after the local heating, 1.25 mM pinacidil (KATP channel opener) and subsequently 25 mM sodium nitroprusside (NO donor) were administered to three of the four sites (each 25-30 min). The local heating-induced prolonged elevation in CVC was attenuated by glibenclamide (19%), but the transient initial peak was not. However, glibenclamide had no effect on the prolonged elevation in CVC in the presence of NOS inhibition. Pinacidil caused an elevation in CVC, but this response was abolished at the glibenclamide-treated skin site, demonstrating its effectiveness as a KATP channel blocker. The pinacidil-induced increase in CVC was unaffected by NOS inhibition, whereas the increase in CVC elicited by sodium nitroprusside was partly (15%) inhibited by glibenclamide. In summary, we showed an interactive effect of KATP channels and NOS for the plateau of cutaneous thermal hyperemia. This interplay may reflect a vascular smooth muscle cell KATP channel activation by NO.


Hyperemia/enzymology , KATP Channels/metabolism , Microcirculation , Microvessels/enzymology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Skin/blood supply , Vasodilation , Adult , Blood Flow Velocity , Enzyme Inhibitors/administration & dosage , Female , Humans , Hyperemia/etiology , Hyperemia/physiopathology , Hypothermia, Induced , Ion Channel Gating , KATP Channels/antagonists & inhibitors , Male , Microcirculation/drug effects , Microvessels/drug effects , Microvessels/physiopathology , Nitric Oxide Donors/administration & dosage , Nitric Oxide Synthase/antagonists & inhibitors , Potassium Channel Blockers/administration & dosage , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/administration & dosage , Young Adult
16.
Brain Behav Immun ; 84: 115-131, 2020 02.
Article En | MEDLINE | ID: mdl-31778743

Sepsis is a host response to systemic inflammation and infection that may lead to multi-organ dysfunction and eventual death. While acute brain dysfunction is common among all sepsis patients, chronic neurological impairment is prevalent among sepsis survivors. The brain microvasculature has emerged as a major determinant of sepsis-associated brain dysfunction, yet the mechanisms that underlie its associated neuroimmune perturbations and behavioral deficits are not well understood. An emerging body of data suggests that inhibition of tissue-nonspecific alkaline phosphatase (TNAP) enzyme activity in cerebral microvessels may be associated with changes in endothelial cell barrier integrity. The objective of this study was to elucidate the connection between alterations in cerebrovascular TNAP enzyme activity and brain microvascular dysfunction in late sepsis. We hypothesized that the disruption of TNAP enzymatic activity in cerebral microvessels would be coupled to the sustained loss of brain microvascular integrity, elevated neuroinflammatory responses, and behavioral deficits. Male mice were subjected to cecal ligation and puncture (CLP), a model of experimental sepsis, and assessed up to seven days post-sepsis. All mice were observed daily for sickness behavior and underwent behavioral testing. Our results showed a significant decrease in brain microvascular TNAP enzyme activity in the somatosensory cortex and spinal cord of septic mice but not in the CA1 and CA3 hippocampal regions. Furthermore, we showed that loss of cerebrovascular TNAP enzyme activity was coupled to a loss of claudin-5 and increased perivascular IgG infiltration in the somatosensory cortex. Analyses of whole brain myeloid and T-lymphoid cell populations also revealed a persistent elevation of infiltrating leukocytes, which included both neutrophil and monocyte myeloid derived suppressor cells (MDSCs). Regional analyses of the somatosensory cortex, hippocampus, and spinal cord revealed significant astrogliosis and microgliosis in the cortex and spinal cord of septic mice that was accompanied by significant microgliosis in the CA1 and CA3 hippocampal regions. Assessment of behavioral deficits revealed no changes in learning and memory or evoked locomotion. However, the hot plate test uncovered a novel anti-nociceptive phenotype in our septic mice, and we speculate that this phenotype may be a consequence of sustained GFAP astrogliosis and loss of TNAP activity in the somatosensory cortex and spinal cord of septic mice. Taken together, these results demonstrate that the loss of TNAP enzyme activity in cerebral microvessels during late sepsis is coupled to sustained neuroimmune dysfunction which may underlie, in part, the chronic neurological impairments observed in sepsis survivors.


Alkaline Phosphatase/metabolism , Brain/blood supply , Inflammation/complications , Inflammation/enzymology , Microvessels/enzymology , Sepsis/complications , Sepsis/psychology , Animals , Brain/pathology , Brain/physiopathology , Cell Line , Disease Models, Animal , Humans , Inflammation/psychology , Male , Mice , Mice, Inbred C57BL , Sepsis/enzymology
17.
Microvasc Res ; 128: 103941, 2020 03.
Article En | MEDLINE | ID: mdl-31678362

BACKGROUND: Activation of the proliferation of pulmonary microvascular endothelial cells (PMVECs) is a key step in the recovery of the integrity of endothelial monolayer, which helps to alleviate acute lung injury (ALI). Platelet endothelial aggregation receptor-1 (PEAR1), expressed on endothelial cells, was reported to inhibit the proliferation of vascular endothelial cells and angiogenesis. However, little is known about its role and mechanism in vascular endothelial disorders in ALI. OBJECTIVE: The aim of this study was to investigate the impact of PEAR1 on the proliferation of pulmonary microvascular endothelial cells in ALI. METHODS: We tested the expression level of PEAR1 in the lungs of WT mice in ALI model induced by intestinal IR. Primary human pulmonary microvascular endothelial cells (HPMECs) were stimulated by 1 mg/L LPS in vitro. We synthesized siPEAR1 and Flag-PEAR1 plasmid to verify the role of PEAR1 on regulating the proliferation of HPMECs under LPS condition and to explore related signaling pathways. RESULTS: The expression level of PEAR1 significantly increased in ALI induced by intestinal IR. PEAR1 knockdown enhanced the proliferation level of HPMECs, which, however, was inhibited by PEAR1 overexpression. PEAR1 knockdown activated PI3K/AKT pathway both in steady state and under LPS condition. PI3K inhibitor, LY294002, reversed the increasing proliferation level and cell progression of HPMECs induced by PEAR1 knockdown after LPS challenge. CONCLUSIONS: PEAR1 acts as a negative regulator in the proliferation of HPMECs in ALI model via the PI3K/AKT pathway.


Acute Lung Injury/enzymology , Cell Proliferation , Endothelial Cells/enzymology , Lung/blood supply , Microvessels/enzymology , Receptors, Cell Surface/metabolism , Acute Lung Injury/pathology , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Humans , Male , Mice, Inbred C57BL , Microvessels/pathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/genetics , Signal Transduction
18.
J Am Heart Assoc ; 8(23): e013130, 2019 12 03.
Article En | MEDLINE | ID: mdl-31752638

Background Uric acid (UA) is a plasmatic antioxidant that has possible effects on blood pressure. The effects of UA on endothelial function are unclear. We hypothesize that endothelial function is not impaired unless significant UA depletion is achieved through selective xanthine oxidase inhibition with febuxostat and recombinant uricase (rasburicase). Methods and Results Microvascular hyperemia, induced by iontophoresis of acetylcholine and sodium nitroprusside, and heating-induced local hyperemia after iontophoresis of saline and a specific nitric oxide synthase inhibitor were assessed by laser Doppler imaging. Blood pressure and renin-angiotensin system markers were measured, and arterial stiffness was assessed. CRP (C-reactive protein), allantoin, chlorotyrosine/tyrosine ratio, homocitrulline/lysine ratio, myeloperoxidase activity, malondialdehyde, and interleukin-8 were used to characterize inflammation and oxidative stress. Seventeen young healthy men were enrolled in a randomized, double-blind, placebo-controlled, 3-way crossover study. The 3 compared conditions were placebo, febuxostat alone, and febuxostat together with rasburicase. The allantoin (µmol/L)/UA (µmol/L) ratio differed between sessions (P<0.0001). During the febuxostat-rasburicase session, heating-induced hyperemia became altered in the presence of nitric oxide synthase inhibition; and systolic blood pressure, angiotensin II, and myeloperoxidase activity decreased (P≤0.03 versus febuxostat). The aldosterone concentration decreased in the febuxostat-rasburicase group (P=0.01). Malondialdehyde increased when UA concentration decreased (both P<0.01 for febuxostat and febuxostat-rasburicase versus placebo). Other parameters remained unchanged. Conclusions A large and short-term decrease in UA in humans alters heat-induced endothelium-dependent microvascular vasodilation, slightly reduces systolic blood pressure through renin-angiotensin system activity reduction, and markedly reduces myeloperoxidase activity when compared with moderate UA reduction. A moderate or severe hypouricemia leads to an increase in lipid peroxidation through loss of antioxidant capacity of plasma. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT03395977.


Blood Pressure/physiology , Endothelium, Vascular/physiopathology , Febuxostat/pharmacology , Microvessels/physiopathology , Urate Oxidase/pharmacology , Uric Acid/blood , Vasodilation/physiology , Xanthine Oxidase/antagonists & inhibitors , Adult , Cross-Over Studies , Double-Blind Method , Endothelium, Vascular/enzymology , Humans , Male , Microvessels/enzymology , Severity of Illness Index , Young Adult
19.
Microvasc Res ; 126: 103891, 2019 11.
Article En | MEDLINE | ID: mdl-31283926

Angiogenesis after cerebral ischemia plays a pivotal role in neurological recovery and represents a therapeutic target. The angiogenic effect of nuclear factor E2-related factor 2 (Nrf2) was identified in recent years. However, the effects of tert-butylhydroquinone, an Nrf2 inducer, on angiogenesis and astrocyte activation after stroke remain unclear. In this study, we investigated whether tert-butylhydroquinone enhanced angiogenesis and astrocyte activation through Nrf2 pathway. Wild-type (Nrf2+/+) and Nrf2-deficient (Nrf2-/-) mice were subjected to permanent distal middle cerebral artery occlusion (dMCAO). We established 6 experimental groups (sham Nrf2+/+ group, vehicle Nrf2+/+ group, tBHQ Nrf2+/+ group; sham Nrf2-/- group, vehicle Nrf2-/- group, and tBHQ Nrf2-/- group). The infarct volume, neurological function, microvessel density (MVD), astrocytic endfeet covered ratio and the expression of Nrf2, HO-1 and VEGF in the ischemic brain were measured at different time points. Compared with that observed in the vehicle Nrf2+/+ group, tBHQ significantly reduced the infarct volume, enhanced post-stroke angiogenesis and astrocytic endfeet covered ratio in the peri-infarct area. The Nrf2/HO-1/VEGF pathway was activated by tBHQ in the angiogenesis process. However, in Nrf2-/- mice, Nrf2 deficiency blocked the effects of tBHQ on angiogenesis process and neurological recovery as well as abolished the mediation of proangiogenic factors. These results suggested that tBHQ enhanced angiogenesis and astrocyte activation through activating Nrf2 pathway after cerebral ischemia.


Angiogenesis Inducing Agents/pharmacology , Astrocytes/drug effects , Brain Ischemia/drug therapy , Brain/blood supply , Heme Oxygenase-1/metabolism , Hydroquinones/pharmacology , Membrane Proteins/metabolism , Microvessels/drug effects , NF-E2-Related Factor 2/metabolism , Neovascularization, Physiologic/drug effects , Animals , Astrocytes/enzymology , Astrocytes/pathology , Brain Ischemia/enzymology , Brain Ischemia/genetics , Brain Ischemia/physiopathology , Cell Proliferation/drug effects , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Male , Mice, Inbred ICR , Mice, Knockout , Microvessels/enzymology , Microvessels/physiopathology , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
20.
Sci Rep ; 9(1): 2567, 2019 02 22.
Article En | MEDLINE | ID: mdl-30796241

Cocaine use is associated with breach in the blood brain barrier (BBB) and increased HIV-1 neuro-invasion. We show that the cellular enzyme "Prolidase" plays a key role in cocaine-induced disruption of the BBB. We established a barrier model to mimic the BBB by culturing human brain microvascular endothelial cells (HBMECs) in transwell inserts. In this model, cocaine treatment enhanced permeability of FITC-dextran suggesting a breach in the barrier. Interestingly, cocaine treatment increased the activity of matrix metallo-proteinases that initiate degradation of the BBB-associated collagen. Cocaine exposure also induced prolidase expression and activity in HBMECs. Prolidase catalyzes the final and rate-limiting step of collagen degradation during BBB remodeling. Knock-down of prolidase abrogated cocaine-mediated increased permeability suggesting a direct role of prolidase in BBB breach. To decipher the mechanism by which cocaine regulates prolidase, we probed the inducible nitric oxide synthase (iNOS) mediated phosphorylation of prolidase since mRNA levels of the protein were not altered upon cocaine treatment. We observed increased iNOS expression concurrent with increased prolidase phosphorylation in cocaine treated cells. Subsequently, inhibition of iNOS decreased prolidase phosphorylation and reduced cocaine-mediated permeability. Finally, cocaine treatment increased transmigration of monocytic cells through the HBMEC barrier. Knock-down of prolidase reduced cocaine-mediated monocyte transmigration, establishing a key role of prolidase in cocaine-induced breach in endothelial cell barrier.


Blood-Brain Barrier/enzymology , Cocaine/adverse effects , Dipeptidases/biosynthesis , Endothelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Microvessels/enzymology , Blood-Brain Barrier/injuries , Blood-Brain Barrier/pathology , Cocaine/pharmacology , Endothelial Cells/pathology , Humans , Microvessels/injuries , Microvessels/pathology , Monocytes/metabolism , Monocytes/pathology , Nitric Oxide Synthase Type II/biosynthesis , THP-1 Cells , Transendothelial and Transepithelial Migration/drug effects
...