Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 44(5): 1124-1134, 2024 May.
Article in English | MEDLINE | ID: mdl-38511328

ABSTRACT

BACKGROUND: SARS-CoV-2 infections cause COVID-19 and are associated with inflammation, coagulopathy, and high incidence of thrombosis. Myeloid cells help coordinate the initial immune response in COVID-19. Although we appreciate that myeloid cells lie at the nexus of inflammation and thrombosis, the mechanisms that unite the two in COVID-19 remain largely unknown. METHODS: In this study, we used systems biology approaches including proteomics, transcriptomics, and mass cytometry to define the circulating proteome and circulating immune cell phenotypes in subjects with COVID-19. RESULTS: In a cohort of subjects with COVID-19 (n=35), circulating markers of inflammation (CCL23 [C-C motif chemokine ligand 23] and IL [interleukin]-6) and vascular dysfunction (ACE2 [angiotensin-converting enzyme 2] and TF [tissue factor]) were elevated in subjects with severe compared with mild COVID-19. Additionally, although the total white blood cell counts were similar between COVID-19 groups, CD14+ (cluster of differentiation) monocytes from subjects with severe COVID-19 expressed more TF. At baseline, transcriptomics demonstrated increased IL-6, CCL3, ACOD1 (aconitate decarboxylase 1), C5AR1 (complement component 5a receptor), C5AR2, and TF in subjects with severe COVID-19 compared with controls. Using stress transcriptomics, we found that circulating immune cells from subjects with severe COVID-19 had evidence of profound immune paralysis with greatly reduced transcriptional activation and release of inflammatory markers in response to TLR (Toll-like receptor) activation. Finally, sera from subjects with severe (but not mild) COVID-19 activated human monocytes and induced TF expression. CONCLUSIONS: Taken together, these observations further elucidate the pathological mechanisms that underlie immune dysfunction and coagulation abnormalities in COVID-19, contributing to our growing understanding of SARS-CoV-2 infections that could also be leveraged to develop novel diagnostic and therapeutic strategies.


Subject(s)
COVID-19 , Monocytes , Thromboplastin , Thrombosis , Adult , Aged , Female , Humans , Male , Middle Aged , Biomarkers/blood , COVID-19/immunology , COVID-19/blood , COVID-19/complications , Monocytes/immunology , Monocytes/metabolism , Proteomics/methods , SARS-CoV-2/physiology , Thromboplastin/metabolism , Thromboplastin/genetics , Thrombosis/immunology , Thrombosis/blood , Thrombosis/etiology
2.
PLoS Comput Biol ; 17(5): e1008861, 2021 05.
Article in English | MEDLINE | ID: mdl-33956786

ABSTRACT

The relationship between regional variabilities in airflow (ventilation) and blood flow (perfusion) is a critical determinant of gas exchange efficiency in the lungs. Hypoxic pulmonary vasoconstriction is understood to be the primary active regulator of ventilation-perfusion matching, where upstream arterioles constrict to direct blood flow away from areas that have low oxygen supply. However, it is not understood how the integrated action of hypoxic pulmonary vasoconstriction affects oxygen transport at the system level. In this study we develop, and make functional predictions with a multi-scale multi-physics model of ventilation-perfusion matching governed by the mechanism of hypoxic pulmonary vasoconstriction. Our model consists of (a) morphometrically realistic 2D pulmonary vascular networks to the level of large arterioles and venules; (b) a tileable lumped-parameter model of vascular fluid and wall mechanics that accounts for the influence of alveolar pressure; (c) oxygen transport accounting for oxygen bound to hemoglobin and dissolved in plasma; and (d) a novel empirical model of hypoxic pulmonary vasoconstriction. Our model simulations predict that under the artificial test condition of a uniform ventilation distribution (1) hypoxic pulmonary vasoconstriction matches perfusion to ventilation; (2) hypoxic pulmonary vasoconstriction homogenizes regional alveolar-capillary oxygen flux; and (3) hypoxic pulmonary vasoconstriction increases whole-lobe oxygen uptake by improving ventilation-perfusion matching.


Subject(s)
Hypoxia/physiopathology , Models, Biological , Pulmonary Circulation/physiology , Ventilation-Perfusion Ratio/physiology , Algorithms , Animals , Arterioles/physiopathology , Biophysical Phenomena , Computational Biology , Computer Simulation , Humans , Lung/blood supply , Lung/physiopathology , Oxygen/physiology , Pulmonary Gas Exchange/physiology , Rats , Vasoconstriction/physiology , Venules/physiopathology
3.
Article in English | MEDLINE | ID: mdl-36260206

ABSTRACT

PURPOSE: Acute heart failure (AHF) syndromes manifest increased inflammation and vascular dysfunction; however, mechanisms that integrate the two in AHF remain largely unknown. The glycocalyx (GAC) is a sugar-based shell that envelops all mammalian cells. Much GAC research has focused on its role in vascular responses, with comparatively little known about how the GAC regulates immune cell function. METHODS: In this study, we sought to determine if GAC degradation products are elevated in AHF patients, how these degradation products relate to circulating inflammatory mediators, and whether the monocyte GAC (mGAC) itself modulates monocyte activation. Inflammatory markers and GAC degradation products were profiled using ELISAs. Flow cytometry was used to assess the mGAC and RNA-seq was employed to understand the role of the mGAC in regulating inflammatory activation programs. RESULTS: In a cohort of hospitalized AHF patients (n = 17), we found that (1) the GAC degradation product heparan sulfate (HS) was elevated compared with age-matched controls (4396 and 2903 ng/mL; p = 0.01) and that (2) HS and soluble CD14 (a marker of monocyte activation) levels were closely related (Pearson's r = 0.65; p = 0.002). Mechanistically, Toll-like receptor (TLR) activation of human monocytes results in GAC remodeling and a decrease in the mGAC (71% compared with no treatment; p = 0.0007). Additionally, we found that ex vivo enzymatic removal of HS and disruption of the mGAC triggers human monocyte activation and amplifies monocyte inflammatory responses. Specifically, using RNA-seq, we found that enzymatic degradation of the mGAC increases transcription of inflammatory (IL6, CCL3) and vascular (tissue factor/F3) mediators. CONCLUSION: These studies indicate that the mGAC is dynamically remodeled during monocyte activation and that mGAC remodeling itself may contribute to the heightened inflammation associated with AHF.

4.
Arterioscler Thromb Vasc Biol ; 40(1): 61-71, 2020 01.
Article in English | MEDLINE | ID: mdl-31619062

ABSTRACT

OBJECTIVE: CD73 is an ectonucleotidase which catalyzes the conversion of AMP (adenosine monophosphate) to adenosine. Adenosine has been shown to be anti-inflammatory and vasorelaxant. The impact of ectonucleotidases on age-dependent atherosclerosis remains unclear. Our aim was to investigate the role of CD73 in age-dependent accumulation of atherosclerosis. Approach and results: Mice doubly deficient in CD73 and ApoE (apolipoprotein E; (cd73-/-/apoE-/-) were generated, and the extent of aortic atherosclerotic plaque was compared with apoE-/- controls at 12, 20, 32, and 52 weeks. By 12 weeks of age, cd73-/-/apoE-/- mice exhibited a significant increase in plaque (1.4±0.5% of the total vessel surface versus 0.4±0.1% in apoE-/- controls, P<0.005). By 20 weeks of age, this difference disappeared (2.9±0.4% versus 3.3±0.7%). A significant reversal in phenotype emerged at 32 weeks (9.8±1.2% versus 18.3±1.4%; P<0.0001) and persisted at the 52 week timepoint (22.4±2.1% versus 37.0±2.1%; P<0.0001). The inflammatory response to aging was found to be comparable between cd73-/-/apoE-/- mice and apoE-/- controls. A reduction in lipolysis in CD73 competent mice was observed, even with similar plasma lipid levels (cd73-/-/apoE-/- versus apoE-/- at 12 weeks [16.2±0.7 versus 9.5±1.4 nmol glycerol/well], 32 weeks [24.1±1.5 versus 7.4±0.4 nmol/well], and 52 weeks [13.8±0.62 versus 12.7±2.0 nmol/well], P<0.001). CONCLUSIONS: At early time points, CD73 exerts a subtle antiatherosclerotic influence, but with age, the pattern reverses, and the presence of CD73 promoted suppression of lipid catabolism.


Subject(s)
5'-Nucleotidase/genetics , Atherosclerosis/genetics , Gene Expression Regulation, Developmental , RNA/genetics , 5'-Nucleotidase/biosynthesis , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
5.
Arterioscler Thromb Vasc Biol ; 39(4): e118-e129, 2019 04.
Article in English | MEDLINE | ID: mdl-30816804

ABSTRACT

Objective- Leukocyte flux contributes to thrombus formation in deep veins under pathological conditions, but mechanisms that inhibit venous thrombosis are incompletely understood. Ectonucleotide di(tri)phosphohydrolase 1 ( ENTPD1 or Cd39), an ectoenzyme that catabolizes extracellular adenine nucleotides, is embedded on the surface of endothelial cells and leukocytes. We hypothesized that under venous stasis conditions, CD39 regulates inflammation at the vein:blood interface in a murine model of deep vein thrombosis. Approach and Results- CD39-null mice developed significantly larger venous thrombi under venous stasis, with more leukocyte recruitment compared with wild-type mice. Gene expression profiling of wild-type and Cd39-null mice revealed 76 differentially expressed inflammatory genes that were significantly upregulated in Cd39-deleted mice after venous thrombosis, and validation experiments confirmed high expression of several key inflammatory mediators. P-selectin, known to have proximal involvement in venous inflammatory and thrombotic events, was upregulated in Cd39-null mice. Inferior vena caval ligation resulted in thrombosis and a corresponding increase in both P-selectin and VWF (von Willebrand Factor) levels which were strikingly higher in mice lacking the Cd39 gene. These mice also manifest an increase in circulating platelet-leukocyte heteroaggregates suggesting heterotypic crosstalk between coagulation and inflammatory systems, which is amplified in the absence of CD39. Conclusions- These data suggest that CD39 mitigates the venous thromboinflammatory response to flow interruption.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , Chemotaxis, Leukocyte/physiology , Hemorheology , Vasculitis/enzymology , Venous Thrombosis/enzymology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Apyrase/deficiency , Apyrase/genetics , Blood Platelets/physiology , Cell Adhesion , Gene Expression Regulation , Gene Regulatory Networks , Ligation , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/biosynthesis , P-Selectin/genetics , Receptors, Purinergic P2Y1/metabolism , Vasculitis/physiopathology , Vena Cava, Inferior , Venous Thrombosis/physiopathology , von Willebrand Factor/biosynthesis , von Willebrand Factor/genetics
6.
Circulation ; 135(24): 2389-2402, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28377485

ABSTRACT

BACKGROUND: Cerebral tissue damage after an ischemic event can be exacerbated by inflammation and thrombosis. Elevated extracellular ATP and ADP levels are associated with cellular injury, inflammation, and thrombosis. Ectonucleoside triphosphate diphosphohydrolase-1 (CD39), an enzyme expressed on the plasmalemma of leukocytes and endothelial cells, suppresses platelet activation and leukocyte infiltration by phosphohydrolyzing ATP/ADP. To investigate the effects of increased CD39 in an in vivo cerebral ischemia model, we developed a transgenic mouse expressing human CD39 (hCD39). METHODS: A floxed-stop sequence was inserted between the promoter and the hCD39 transcriptional start site, generating a mouse in which the expression of hCD39 can be controlled tissue-specifically using Cre recombinase mice. We generated mice that express hCD39 globally or in myeloid-lineage cells only. Cerebral ischemia was induced by middle cerebral artery occlusion. Infarct volumes were quantified by MRI after 48 hours. RESULTS: Both global and transgenic hCD39- and myeloid lineage CD39-overexpressing mice (transgenic, n=9; myeloid lineage, n=6) demonstrated significantly smaller cerebral infarct volumes compared with wild-type mice. Leukocytes from ischemic and contralateral hemispheres were analyzed by flow cytometry. Although contralateral hemispheres had equal numbers of macrophages and neutrophils, ischemic hemispheres from transgenic mice had less infiltration (n=4). Transgenic mice showed less neurological deficit compared with wild-type mice (n=6). CONCLUSIONS: This is the first report of transgenic overexpression of CD39 in mice imparting a protective phenotype after stroke, with reduced leukocyte infiltration, smaller infarct volumes, and decreased neurological deficit. CD39 overexpression, either globally or in myeloid lineage cells, quenches postischemic leukosequestration and reduces stroke-induced neurological injury.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Apyrase/biosynthesis , Apyrase/genetics , Brain Ischemia/genetics , Brain Ischemia/metabolism , Cell Lineage/physiology , Transgenes/physiology , Animals , Brain Ischemia/prevention & control , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/physiology
7.
Circ Res ; 119(1): 142-58, 2016 06 24.
Article in English | MEDLINE | ID: mdl-27340273

ABSTRACT

Over the past several decades, there have been substantial advances in our knowledge of the pathophysiology of stroke. Understanding the benefits of timely reperfusion has led to the development of thrombolytic therapy as the cornerstone of current management of ischemic stroke, but there remains much to be learned about mechanisms of neuronal ischemic and reperfusion injury and associated inflammation. For ischemic stroke, novel therapeutic targets have continued to remain elusive. When considering modern molecular biological techniques, advanced translational stroke models, and clinical studies, a consistent pattern emerges, implicating perturbation of the immune equilibrium by stroke in both central nervous system injury and repair responses. Stroke triggers activation of the neuroimmune axis, comprised of multiple cellular constituents of the immune system resident within the parenchyma of the brain, leptomeninges, and vascular beds, as well as through secretion of biological response modifiers and recruitment of immune effector cells. This neuroimmune activation can directly impact the initiation, propagation, and resolution phases of ischemic brain injury. To leverage a potential opportunity to modulate local and systemic immune responses to favorably affect the stroke disease curve, it is necessary to expand our mechanistic understanding of the neuroimmune axis in ischemic stroke. This review explores the frontiers of current knowledge of innate and adaptive immune responses in the brain and how these responses together shape the course of ischemic stroke.


Subject(s)
Neuroimmunomodulation , Stroke/immunology , Adaptive Immunity , Animals , Blood-Brain Barrier/metabolism , Humans , Immunity, Innate , Stroke/metabolism
8.
Trans Am Clin Climatol Assoc ; 129: 132-139, 2018.
Article in English | MEDLINE | ID: mdl-30166707

ABSTRACT

A common thread underlying vascular or tissue injury is the loss of plasmalemmal integrity and the passive (or even active) spillage of intracellular contents into the circulation. Purinergic nucleotides, which serve as energy shuttling moieties within cells, are among the contents released into the bloodstream, where they signal danger and trigger thrombosis and inflammation. To regain vascular homeostasis, vascular cells have evolved highly conserved mechanisms to transact the catalytic degradation of extracellular nucleotides such as adenosine triphosphate (ATP) and adenosine diphosphate (ADP). CD39, the main endothelial ectonucleotidase which cleaves ATP and ADP, plays an essential role in ridding the bloodstream of these danger signals, thereby sustaining vascular homeostasis. Studies herein describe the upregulation of endothelial CD39 gene by steady laminar shear forces, and conversely, its downregulation under turbulent flow conditions. CD39 appears to be a critical ectonucleotidase which suppresses atherogenesis under experimental hyperlipidemic conditions in mice, and which also significantly mitigates pathologic vascular remodeling and development of pulmonary arterial hypertension in mice placed under chronic hypoxic conditions. Together, these data reveal that CD39 opposes pathologic vascular remodeling under hyperlipidemic or hypoxic conditions. CD39 can therefore be viewed as a critical vascular homeostatic regulator to sustain vascular quiescence and to protect against pathological vascular remodeling in diseases as diverse as atherosclerosis and pulmonary arterial hypertension.


Subject(s)
Apyrase/metabolism , Atherosclerosis/enzymology , Hypertension, Pulmonary/enzymology , Pulmonary Artery/enzymology , Vascular Remodeling , Animals , Apyrase/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Mechanotransduction, Cellular , Plaque, Atherosclerotic , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Regional Blood Flow , Signal Transduction
9.
Bioorg Med Chem ; 25(16): 4487-4496, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28705434

ABSTRACT

The surface properties of nanoparticles (NPs) are a major factor that influences how these nanomaterials interact with biological systems. Interactions between NPs and macrophages of the reticuloendothelial system (RES) can reduce the efficacy of NP diagnostics and therapeutics. Traditionally, to limit NP clearance by the RES system, the NP surface is neutralized with molecules like poly(ethylene glycol) (PEG) which are known to resist protein adsorption and RES clearance. Unfortunately, PEG modification is not without drawbacks including difficulties with the synthesis and associations with immune reactions. To overcome some of these obstacles, we neutralized the NP surface by acetylation and compared this modification to PEGylation for RES clearance and tumor-specific targeting. We found that acetylation was comparable to PEGylation in reducing RES clearance. Additionally, we found that dendrimer acetylation did not impact folic acid (FA)-mediated targeting of tumor cells whereas PEG surface modification reduced the targeting ability of the NP. These results clarify the impact of different NP surface modifications on RES clearance and cell-specific targeting and provide insights into the design of more effective NPs.


Subject(s)
Folic Acid/pharmacology , Macrophages/chemistry , Nanoparticles/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Folic Acid/chemistry , Humans , KB Cells , Mice , Molecular Structure , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , RAW 264.7 Cells , Structure-Activity Relationship , Surface Properties
10.
Am J Physiol Heart Circ Physiol ; 311(1): H286-98, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27208163

ABSTRACT

Despite the fact that nucleotides and adenosine help regulate vascular tone through purinergic signaling pathways, little is known regarding their contributions to the pathobiology of pulmonary arterial hypertension, a condition characterized by elevated pulmonary vascular resistance and remodeling. Even less is known about the potential role that alterations in CD39 (ENTPD1), the ectonucleotidase responsible for the conversion of the nucleotides ATP and ADP to AMP, may play in pulmonary arterial hypertension. In this study we identified decreased CD39 expression on the pulmonary endothelium of patients with idiopathic pulmonary arterial hypertension. We next determined the effects of CD39 gene deletion in mice exposed to normoxia or normobaric hypoxia (10% oxygen). Compared with controls, hypoxic CD39(-/-) mice were found to have a markedly elevated ATP-to-adenosine ratio, higher pulmonary arterial pressures, more right ventricular hypertrophy, more arterial medial hypertrophy, and a pro-thrombotic phenotype. In addition, hypoxic CD39(-/-) mice exhibited a marked increase in lung P2X1 receptors. Systemic reconstitution of ATPase and ADPase enzymatic activities through continuous administration of apyrase decreased pulmonary arterial pressures in hypoxic CD39(-/-) mice to levels found in hypoxic CD39(+/+) controls. Treatment with NF279, a potent and selective P2X1 receptor antagonist, lowered pulmonary arterial pressures even further. Our study is the first to implicate decreased CD39 and resultant alterations in circulating purinergic signaling ligands and cognate receptors in the pathobiology of pulmonary arterial hypertension. Reconstitution and receptor blocking experiments suggest that phosphohydrolysis of purinergic nucleotide tri- and diphosphates, or blocking of the P2X1 receptor could serve as treatment for pulmonary arterial hypertension.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Hypertension, Pulmonary/metabolism , Lung/metabolism , Pulmonary Artery/metabolism , Receptors, Purinergic P2X1/metabolism , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Antihypertensive Agents/pharmacology , Apyrase/deficiency , Apyrase/genetics , Apyrase/pharmacology , Arterial Pressure , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Hydrolysis , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Lung/drug effects , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X1/drug effects , Severity of Illness Index , Signal Transduction , Suramin/analogs & derivatives , Suramin/pharmacology , Vascular Remodeling , Ventricular Remodeling
11.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L749-63, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24532288

ABSTRACT

Bronchopulmonary dysplasia (BPD), a lung disease of prematurely born infants, is characterized in part by arrested development of pulmonary alveolae. We hypothesized that heme oxygenase (HO-1) and its byproduct carbon monoxide (CO), which are thought to be cytoprotective against redox stress, mitigate lung injury and alveolar simplification in hyperoxia-exposed neonatal mice, a model of BPD. Three-day-old C57BL/6J mice were exposed to air or hyperoxia (FiO2, 75%) in the presence or absence of inhaled CO (250 ppm for 1 h twice daily) for 21 days. Hyperoxic exposure increased mean linear intercept, a measure of alveolar simplification, whereas CO treatment attenuated hypoalveolarization, yielding a normal-appearing lung. Conversely, HO-1-null mice showed exaggerated hyperoxia-induced hypoalveolarization. CO also inhibited hyperoxia-induced pulmonary accumulation of F4/80+, CD11c+, and CD11b+ monocytes and Gr-1+ neutrophils. Furthermore, CO attenuated lung mRNA and protein expression of proinflammatory cytokines, including the monocyte chemoattractant CCL2 in vivo, and decreased hyperoxia-induced type I alveolar epithelial cell CCL2 production in vitro. Hyperoxia-exposed CCL2-null mice, like CO-treated mice, showed attenuated alveolar simplification and lung infiltration of CD11b+ monocytes, consistent with the notion that CO blocks lung epithelial cell cytokine production. We conclude that, in hyperoxia-exposed neonatal mice, inhalation of CO suppresses inflammation and alveolar simplification.


Subject(s)
Antimetabolites/pharmacology , Carbon Monoxide/pharmacology , Chemokine CCL2/physiology , Heme Oxygenase-1/metabolism , Hyperoxia/physiopathology , Pneumonia/drug therapy , Pulmonary Alveoli/drug effects , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Heme Oxygenase-1/genetics , Hyperoxia/drug therapy , Immunoenzyme Techniques , Macrophages, Alveolar , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes , Oxygen/metabolism , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
Curr Atheroscler Rep ; 16(7): 425, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24838375

ABSTRACT

Extracellular nucleotides play a critical role in vascular thrombosis and inflammation. Alterations in purinergic extracellular nucleotide concentrations activate pathways that result in platelet degranulation and aggregation, and endothelial and leukocyte activation and recruitment. CD39, the dominant vascular nucleotidase, hydrolyzes ATP and ADP to provide the substrate for generation of the anti-inflammatory and antithrombotic mediator adenosine. The purinergic signaling system, with CD39 at its center, plays an important role in modulating vascular homeostasis and the response to vascular injury, as seen in clinically relevant diseases such as stroke, ischemia-reperfusion injury, and pulmonary hypertension. A growing body of knowledge of the purinergic signaling pathway implicates CD39 as a critical modulator of vascular thrombosis and inflammation. Therapeutic strategies targeting CD39 offer promising opportunities in the management of vascular thromboinflammatory diseases.


Subject(s)
Antigens, CD/immunology , Apyrase/immunology , Endothelium, Vascular/immunology , Thrombosis/immunology , Vasculitis/immunology , Antigens, CD/physiology , Apyrase/physiology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Humans , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/metabolism , Inflammation/metabolism , Myocardial Ischemia/immunology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Signal Transduction/immunology , Stroke/immunology , Stroke/metabolism , Thrombosis/metabolism , Vasculitis/metabolism
13.
FASEB J ; 27(11): 4419-28, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23901069

ABSTRACT

The ectoenzyme CD39 suppresses thrombosis and inflammation by suppressing ATP and ADP to AMP. However, mechanisms of CD39 transcriptional and post-translational regulation are not well known. Here we show that CD39 levels are modulated by inhibition of phosphodiesterase 3 (PDE3). RAW macrophages and human umbilical vein endothelial cells (HUVECs) were treated with the PDE3 inhibitors cilostazol and milrinone, then analyzed using qRT-PCR, immunoprecipitation/Western blot, immunofluorescent staining, radio-thin-layer chromatography, a malachite green assay, and ELISA. HUVECs expressed elevated CD39 protein (2-fold [P<0.05] for cilostazol and 2.5-fold [P<0.01] for milrinone), while macrophage CD39 mRNA and protein were both elevated after PDE3 inhibition. HUVEC ATPase activity increased by 25% with cilostazol and milrinone treatment (P<0.05 and P<0.01, respectively), as did ADPase activity (47% and 61%, P<0.001). There was also a dose-dependent elevation of soluble CD39 after treatment with 8-Br-cAMP, with maximal elevation of 60% more CD39 present compared to controls (1 mM, P<0.001). Protein harvested after 8-Br-cAMP treatment showed that ubiquitination of CD39 was decreased by 43% compared to controls. A DMSO or PBS vehicle control was included for each experiment based on solubility of cilostazol, milrinone, and 8-Br-cAMP. These results indicate that PDE3 inhibition regulates endothelial CD39 at a post-translational level.


Subject(s)
Antigens, CD/genetics , Apyrase/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Gene Expression Regulation, Enzymologic , Transcription, Genetic , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cilostazol , Cyclic Nucleotide Phosphodiesterases, Type 3/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/metabolism , Milrinone/pharmacology , Tetrazoles/pharmacology , Ubiquitination
14.
J Immunol ; 188(5): 2387-98, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22291183

ABSTRACT

Ectoenzymes expressed on the surface of vascular cells and leukocytes modulate the ambient nucleotide milieu. CD73 is an ecto-5' nucleotidase that catalyzes the terminal phosphohydrolysis of AMP and resides in the brain on glial cells, cells of the choroid plexus, and leukocytes. Though CD73 tightens epithelial barriers, its role in the ischemic brain remains undefined. When subjected to photothrombotic arterial occlusion, CD73(-/-) mice exhibited significantly larger (49%) cerebral infarct volumes than wild-type mice, with concordant increases in local accumulation of leukocyte subsets (neutrophils, T lymphocytes, macrophages, and microglia). CD73(-/-) mice were rescued from ischemic neurologic injury by soluble 5'-nucleotidase. In situ, CD73(-/-) macrophages upregulated expression of costimulatory molecules far more than wild-type macrophages, with a sharp increase of the CD80/CD86 ratio. To define the CD73-bearing cells responsible for ischemic cerebroprotection, mice were subjected to irradiative myeloablation, marrow reconstitution, and then stroke following engraftment. Chimeric mice lacking CD73 in tissue had larger cerebral infarct volumes and more tissue leukosequestration than did mice lacking CD73 on circulating cells. These data show a cardinal role for CD73 in suppressing ischemic tissue leukosequestration. This underscores a critical role for CD73 as a modulator of brain inflammation and immune function.


Subject(s)
5'-Nucleotidase/physiology , Brain Ischemia/immunology , Brain Ischemia/pathology , Cell Movement/genetics , Cell Movement/immunology , Leukocytes/immunology , Leukocytes/pathology , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Adenosine/biosynthesis , Adenosine/physiology , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Brain Edema/enzymology , Brain Edema/immunology , Brain Edema/pathology , Brain Ischemia/enzymology , Extracellular Fluid/enzymology , Extracellular Fluid/immunology , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Inflammation/enzymology , Inflammation/immunology , Inflammation/prevention & control , Leukocytes/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Tissue Distribution/genetics , Tissue Distribution/immunology
15.
J Immunol ; 189(1): 356-64, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22661086

ABSTRACT

Mechanical ventilation using high oxygen tensions is often necessary to treat patients with respiratory failure. Recently, TLRs were identified as regulators of noninfectious oxidative lung injury. IRAK-M is an inhibitor of MyD88-dependent TLR signaling. Exposure of mice deficient in IRAK-M (IRAK-M(-/-)) to 95% oxygen resulted in reduced mortality compared with wild-type mice and occurred in association with decreased alveolar permeability and cell death. Using a bone marrow chimera model, we determined that IRAK-M's effects were mediated by structural cells rather than bone marrow-derived cells. We confirmed the expression of IRAK-M in alveolar epithelial cells (AECs) and showed that hyperoxia can induce the expression of this protein. In addition, IRAK-M(-/-) AECs exposed to hyperoxia experienced a decrease in cell death. IRAK-M may potentiate hyperoxic injury by suppression of key antioxidant pathways, because lungs and AECs isolated from IRAK-M(-/-) mice have increased expression/activity of heme oxygenase-1, a phase II antioxidant, and NF (erythroid-derived)-related factor-2, a transcription factor that initiates antioxidant generation. Treatment of IRAK-M(-/-) mice in vivo and IRAK-M(-/-) AECs in vitro with the heme oxygenase-1 inhibitor, tin protoporphyrin, substantially decreased survival and significantly reduced the number of live cells after hyperoxia exposure. Collectively, our data suggest that IRAK-M inhibits the induction of antioxidants essential for protecting the lungs against cell death, resulting in enhanced susceptibility to hyperoxic lung injury.


Subject(s)
Acute Lung Injury/immunology , Hyperoxia/immunology , Oxidants/physiology , Pulmonary Alveoli/immunology , Respiratory Mucosa/immunology , Signal Transduction/immunology , Toll-Like Receptors/physiology , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Cell Death/genetics , Cell Death/immunology , Cell Line , Hyperoxia/pathology , Hyperoxia/prevention & control , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/physiology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction/genetics , Toll-Like Receptors/antagonists & inhibitors
16.
Adv Healthc Mater ; : e2400443, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38898728

ABSTRACT

Neutrophils can contribute to inflammatory disease propagation via innate mechanisms intended for inflammation resolution. For example, neutrophil extracellular traps (NETs) are necessary for trapping pathogens but can contribute to clot formation and blood flow restriction, that is, ischemia. Currently, no therapeutics in the clinic directly target NETs despite the known involvement of NETs contributing to mortality and increased disease severity. Vascular-deployed particle-based therapeutics are a novel and robust alternative to traditional small-molecule drugs by enhancing drug delivery to cells of interest. This work designs a high-throughput assay to investigate the immunomodulatory behavior and functionality of salicylic acid-based polymer-based particle therapeutics against NETosis in human neutrophils. Briefly, this work finds that polymeric composition plays a role, and particle size can also influence rates of NETosis. Salicylate-based polymeric (Poly-SA) particles are found to functionally inhibit NETosis depending on the particle size and concentration exposed to neutrophils. This work demonstrates the high throughput method can help fast-track particle-based therapeutic optimization and design, more efficiently preparing this innovative therapeutics for the clinic.

17.
J Heart Lung Transplant ; 42(3): 317-326, 2023 03.
Article in English | MEDLINE | ID: mdl-36522238

ABSTRACT

BACKGROUND: Small airway inflammation and fibrosis or bronchiolitis obliterans (BO) is the predominant presentation of chronic lung allograft dysfunction (CLAD) post-lung transplantation. Carbon monoxide (CO) is a critical endogenous signaling transducer with known anti-inflammatory and anti-fibrotic effects but its therapeutic potential in CLAD remains to be fully elucidated. METHODS: Here we investigate the effect of inhaled CO in modulating chronic lung allograft rejection pathology in a murine orthotopic lung transplant model of BO (B6D2F1/J→DBA/2J). Additionally, the effects of CO on the activated phenotype of mesenchymal cells isolated from human lung transplant recipients with CLAD were studied. RESULTS: Murine lung allografts treated with CO (250 ppm × 30 minutes twice daily from days 7 to 40 post-transplantation) demonstrated decreased immune cell infiltration, fibrosis, and airway obliteration by flow cytometry, trichrome staining, and morphometric analysis, respectively. Decreased total collagen, with levels comparable to isografts, was noted in CO-treated allografts by quantitative hydroxyproline assay. In vitro, CO (250 ppm × 16h) was effective in reversing the fibrotic phenotype of human CLAD mesenchymal cells with decreased collagen I and ß-catenin expression as well as an inhibitory effect on ERK1/2 MAPK, and mTORC1/2 signaling. Sildenafil, a phosphodiesterase 5 inhibitor, partially mimicked the effects of CO on CLAD mesenchymal cells and was partially effective in decreasing collagen deposition in murine allografts, suggesting the contribution of cGMP-dependent and -independent mechanisms in mediating the effect of CO. CONCLUSION: These results suggest a potential role for CO in alleviating allograft fibrosis and mitigating chronic rejection pathology post-lung transplant.


Subject(s)
Bronchiolitis Obliterans , Lung Transplantation , Humans , Animals , Mice , Carbon Monoxide , Allografts/pathology , Lung Transplantation/adverse effects , Fibrosis , Lung/pathology , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/prevention & control , Collagen , Graft Rejection
18.
Nat Commun ; 14(1): 2462, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37117163

ABSTRACT

The combination of inflammation and thrombosis is a hallmark of many cardiovascular diseases. Under such conditions, platelets are recruited to an area of inflammation by forming platelet-leukocyte aggregates via interaction of PSGL-1 on leukocytes and P-selectin on activated platelets, which can bind to the endothelium. While particulate drug carriers have been utilized to passively redirect leukocytes from areas of inflammation, the downstream impact of these carriers on platelet accumulation in thromboinflammatory conditions has yet to be studied. Here, we explore the ability of polymeric particles to divert platelets away from inflamed blood vessels both in vitro and in vivo. We find that untargeted and targeted micron-sized polymeric particles can successfully reduce platelet adhesion to an inflamed endothelial monolayer in vitro in blood flow systems and in vivo in a lipopolysaccharide-induced, systemic inflammation murine model. Our data represent initial work in developing cargo-free, anti-platelet therapeutics specifically for conditions of thromboinflammation.


Subject(s)
Neutrophils , Thrombosis , Humans , Animals , Mice , Neutrophils/metabolism , Inflammation/metabolism , Thromboinflammation , Thrombosis/metabolism , Blood Platelets/metabolism , Leukocytes/metabolism , P-Selectin/metabolism
19.
J Am Heart Assoc ; 12(9): e027920, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37119076

ABSTRACT

Background Ectonucleotidases maintain vascular homeostasis by metabolizing extracellular nucleotides, modulating inflammation and thrombosis, and potentially, myocardial flow through adenosine generation. Evidence implicates dysfunction or deficiency of ectonucleotidases CD39 or CD73 in human disease; the utility of measuring levels of circulating ectonucleotidases as plasma biomarkers of coronary artery dysfunction or disease has not been previously reported. Methods and Results A total of 529 individuals undergoing clinically indicated positron emission tomography stress testing between 2015 and 2019 were enrolled in this single-center retrospective analysis. Baseline demographics, clinical data, nuclear stress test, and coronary artery calcium score variables were collected, as well as a blood sample. CD39 and CD73 levels were assessed as binary (detectable, undetectable) or continuous variables using ELISAs. Plasma CD39 was detectable in 24% of White and 8% of Black study participants (P=0.02). Of the clinical history variables examined, ectonucleotidase levels were most strongly associated with underlying liver disease and not other traditional coronary artery disease risk factors. Intriguingly, detection of circulating ectonucleotidase was inversely associated with stress myocardial blood flow (2.3±0.8 mL/min per g versus 2.7 mL/min per g±1.1 for detectable versus undetectable CD39 levels, P<0.001) and global myocardial flow reserve (Pearson correlation between myocardial flow reserve and log(CD73) -0.19, P<0.001). A subanalysis showed these differences held true independent of liver disease. Conclusions Vasodilatory adenosine is the expected product of local ectonucleotidase activity, yet these data support an inverse relationship between plasma ectonucleotidases, stress myocardial blood flow (CD39), and myocardial flow reserve (CD73). These findings support the conclusion that plasma levels of ectonucleotidases, which may be shed from the endothelial surface, contribute to reduced stress myocardial blood flow and myocardial flow reserve.


Subject(s)
Coronary Artery Disease , Myocardial Perfusion Imaging , Humans , Retrospective Studies , Tomography, X-Ray Computed , Adenosine , Coronary Vessels/diagnostic imaging , Perfusion , Myocardial Perfusion Imaging/methods , Coronary Artery Disease/diagnosis , Coronary Circulation
20.
J Immunol ; 185(2): 1321-9, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20548026

ABSTRACT

There are multiple drivers of leukocyte recruitment in lung allografts that contribute to lymphocytic bronchitis (LB) and bronchiolitis obliterans (BO). The innate mechanisms driving (or inhibiting) leukocyte trafficking to allografts remain incompletely understood. This study tested the hypothesis that CD73 (ecto-5'nucleotidase), an enzyme that catalyzes the conversion of AMP to adenosine, is a critical negative regulator of LB and BO. Implantation of tracheal allografts from wild type (WT) mice into CD73(-/-) recipients revealed a striking increase in airway luminal obliteration at 7 d (62 +/- 4% and 47 +/- 5% for CD73(-/-) and WT allograft recipients, respectively; p = 0.046). There was also a concordant increase in CD3(+) lymphocytic infiltration (523 +/- 41 cells and 313 +/- 43 cells for CD73(-/-) and WT allograft recipients, respectively; p = 0.013). Because real-time PCR revealed a 43-fold upregulation of mRNA for the adenosine A2A receptor (A2AR) in WT allografts compared with WT isografts (p = 0.032), additional experiments were performed to determine whether the protective effect of CD73 was due to generation of adenosine and its stimulation of the A2AR. Treatment of WT recipients with an A2AR agonist significantly reduced CD3(+) lymphocyte infiltration and airway luminal obliteration; similar treatment of CD73(-/-) recipients rescued them from LB and airway obliteration. These data implicate CD73 acting through adenosine generation and its stimulation of the A2AR as a critical negative modulator of lymphocyte recruitment into airway allografts. The CD73/adenosine axis might be a new therapeutic target to prevent BO.


Subject(s)
5'-Nucleotidase/metabolism , Graft Rejection/immunology , Receptor, Adenosine A2A/immunology , Trachea/immunology , 5'-Nucleotidase/genetics , Adenosine/analogs & derivatives , Adenosine/blood , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Animals , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/prevention & control , CD3 Complex/immunology , Chromatography, Liquid , Gene Expression , Graft Rejection/prevention & control , Interferon-gamma/genetics , Interleukin-2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenethylamines/blood , Phenethylamines/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tandem Mass Spectrometry , Trachea/metabolism , Trachea/transplantation , Transplantation, Homologous , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL