Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Am J Pathol ; 191(1): 52-65, 2021 01.
Article in English | MEDLINE | ID: mdl-33069720

ABSTRACT

Endothelial barrier integrity is required for maintaining vascular homeostasis and fluid balance between the circulation and surrounding tissues and for preventing the development of vascular disease. Despite comprehensive understanding of the molecular mechanisms and signaling pathways that mediate endothelial injury, the regulatory mechanisms responsible for endothelial regeneration and vascular repair are incompletely understood and constitute an emerging area of research. Endogenous and exogenous reparative mechanisms serve to reverse vascular damage and restore endothelial barrier function through regeneration of a functional endothelium and re-engagement of endothelial junctions. In this review, mechanisms that contribute to endothelial regeneration and vascular repair are described. Targeting these mechanisms has the potential to improve outcome in diseases that are characterized by vascular injury, such as atherosclerosis, restenosis, peripheral vascular disease, sepsis, and acute respiratory distress syndrome. Future studies to further improve current understanding of the mechanisms that control endothelial regeneration and vascular repair are also highlighted.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Regeneration/physiology , Regenerative Medicine , Animals , Endothelium, Vascular/injuries , Humans
2.
Nature ; 540(7632): 236-241, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27798602

ABSTRACT

R-2-hydroxyglutarate accumulates to millimolar levels in cancer cells with gain-of-function isocitrate dehydrogenase 1/2 mutations. These levels of R-2-hydroxyglutarate affect 2-oxoglutarate-dependent dioxygenases. Both metabolite enantiomers, R- and S-2-hydroxyglutarate, are detectible in healthy individuals, yet their physiological function remains elusive. Here we show that 2-hydroxyglutarate accumulates in mouse CD8+ T cells in response to T-cell receptor triggering, and accumulates to millimolar levels in physiological oxygen conditions through a hypoxia-inducible factor 1-alpha (HIF-1α)-dependent mechanism. S-2-hydroxyglutarate predominates over R-2-hydroxyglutarate in activated T cells, and we demonstrate alterations in markers of CD8+ T-cell differentiation in response to this metabolite. Modulation of histone and DNA demethylation, as well as HIF-1α stability, mediate these effects. S-2-hydroxyglutarate treatment greatly enhances the in vivo proliferation, persistence and anti-tumour capacity of adoptively transferred CD8+ T cells. Thus, S-2-hydroxyglutarate acts as an immunometabolite that links environmental context, through a metabolic-epigenetic axis, to immune fate and function.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Glutarates/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA/chemistry , DNA/metabolism , DNA Methylation/drug effects , Dioxygenases/metabolism , Glutarates/immunology , Glutarates/metabolism , Histones/metabolism , Homeostasis/drug effects , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ketoglutaric Acids/metabolism , Lymphocyte Activation , Lysine/metabolism , Mice , Oxygen/metabolism , Protein Stability , Receptors, Antigen, T-Cell/immunology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
3.
Eur Respir J ; 58(3)2021 09.
Article in English | MEDLINE | ID: mdl-33509961

ABSTRACT

Pulmonary arterial hypertension (PAH) is a devastating disease that involves pulmonary vasoconstriction, small vessel obliteration, large vessel thickening and obstruction, and development of plexiform lesions. PAH vasculopathy leads to progressive increases in pulmonary vascular resistance, right heart failure and, ultimately, premature death. Besides other cell types that are known to be involved in PAH pathogenesis (e.g. smooth muscle cells, fibroblasts and leukocytes), recent studies have demonstrated that endothelial cells (ECs) have a crucial role in the initiation and progression of PAH. The EC-specific role in PAH is multi-faceted and affects numerous pathophysiological processes, including vasoconstriction, inflammation, coagulation, metabolism and oxidative/nitrative stress, as well as cell viability, growth and differentiation. In this review, we describe how EC dysfunction and cell signalling regulate the pathogenesis of PAH. We also highlight areas of research that warrant attention in future studies, and discuss potential molecular signalling pathways in ECs that could be targeted therapeutically in the prevention and treatment of PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Endothelial Cells , Familial Primary Pulmonary Hypertension , Humans , Myocytes, Smooth Muscle , Pulmonary Artery
4.
Am J Pathol ; 189(8): 1664-1679, 2019 08.
Article in English | MEDLINE | ID: mdl-31121134

ABSTRACT

Endothelial barrier dysfunction is a central factor in the pathogenesis of persistent lung inflammation and protein-rich edema formation, the hallmarks of acute respiratory distress syndrome. However, little is known about the molecular mechanisms that are responsible for vascular repair and resolution of inflammatory injury after sepsis challenge. Herein, we show that hypoxia-inducible factor-1α (HIF-1α), expressed in endothelial cells (ECs), is the critical transcriptional factor mediating vascular repair and resolution of inflammatory lung injury. After sepsis challenge, HIF-1α but not HIF-2α expression was rapidly induced in lung vascular ECs, and mice with EC-restricted disruption of Hif1α (Hif1af/f/Tie2Cre+) exhibited defective vascular repair, persistent inflammation, and increased mortality in contrast with the wild-type littermates after polymicrobial sepsis or endotoxemia challenge. Hif1af/f/Tie2Cre+ lungs exhibited marked decrease of EC proliferation during recovery after sepsis challenge, which was associated with inhibited expression of forkhead box protein M1 (Foxm1), a reparative transcription factor. Therapeutic restoration of endothelial Foxm1 expression, via liposomal delivery of Foxm1 plasmid DNA to Hif1af/f/Tie2Cre+ mice, resulted in reactivation of the vascular repair program and improved survival. Together, our studies, for the first time, delineate the essential role of endothelial HIF-1α in driving the vascular repair program. Thus, therapeutic activation of HIF-1α-dependent vascular repair may represent a novel and effective therapy to treat inflammatory vascular diseases, such as sepsis and acute respiratory distress syndrome.


Subject(s)
Endothelial Cells/metabolism , Forkhead Box Protein M1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Injury/metabolism , Lung/physiology , Regeneration , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelial Cells/pathology , Female , Forkhead Box Protein M1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Injury/etiology , Lung Injury/genetics , Lung Injury/pathology , Male , Mice , Mice, Transgenic , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Sepsis/complications , Sepsis/genetics , Sepsis/metabolism , Sepsis/pathology
5.
Thromb J ; 17: 16, 2019.
Article in English | MEDLINE | ID: mdl-31423111

ABSTRACT

Risk factors for thrombosis include hypoxia and sepsis, but the mechanisms that control sepsis-induced thrombus formation are incompletely understood. A recent article published in Thrombosis Journal: (i) reviews the role of endothelial cells in the pathogenesis of sepsis-associated microthrombosis; (ii) describes a novel 'two-path unifying theory' of hemostatic discorders; and (iii) refers to hypoxia as a consequence of microthrombus formation in sepsis patients. The current article adds to this review by describing how sepsis and thrombus formation could be linked through hypoxia and activation of hypoxia-inducible transcription factors (HIFs). In other words, hypoxia and HIF activation may be a cause as well as a consequence of thrombosis in sepsis patients. While microthrombosis reduces microvascular blood flow causing local hypoxia and tissue ischemia, sepsis-induced increases in HIF1 activation could conversely increase the expression of coagulant factors and integrins that promote thrombus formation, and stimulate the formation of pro-thrombotic neutrophil extracellular traps. A better understanding of the role of cell-specific HIFs in thrombus formation could lead to the development of novel prophylactic therapies for individuals at risk of thrombosis.

6.
Am J Respir Crit Care Med ; 198(11): 1423-1434, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29924941

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a devastating disease characterized by progressive vasoconstriction and obliterative vascular remodeling that leads to right heart failure (RHF) and death. Current therapies do not target vascular remodeling and RHF, and result in only modest improvement of morbidity and mortality. OBJECTIVES: To determine whether targeting HIF-2α (hypoxia-inducible factor-2α) with a HIF-2α-selective inhibitor could reverse PAH and RHF in various rodent PAH models. METHODS: HIF-2α and its downstream genes were evaluated in lung samples and pulmonary arterial endothelial cells and smooth muscle cells from patients with idiopathic PAH as well as various rodent PAH models. A HIF-2α-selective inhibitor was used in human lung microvascular endothelial cells and in Egln1Tie2Cre mice, and in Sugen 5416/hypoxia- or monocrotaline-exposed rats. MEASUREMENTS AND MAIN RESULTS: Upregulation of HIF-2α and its target genes was observed in lung tissues and isolated pulmonary arterial endothelial cells from patients with idiopathic PAH and three distinct rodent PAH models. Pharmacological inhibition of HIF-2α by the HIF-2α translation inhibitor C76 (compound 76) reduced right ventricular systolic pressure and right ventricular hypertrophy and inhibited RHF and fibrosis as well as obliterative pulmonary vascular remodeling in Egln1Tie2Cre mice and Sugen 5416/hypoxia PAH rats. Treatment of monocrotaline-exposed PAH rats with C76 also reversed right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary vascular remodeling; prevented RHF; and promoted survival. CONCLUSIONS: These findings demonstrate that pharmacological inhibition of HIF-2α is a promising novel therapeutic strategy for the treatment of severe vascular remodeling and right heart failure in patients with PAH.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Glycolipids/administration & dosage , Heart Failure/physiopathology , Hypertension, Pulmonary/physiopathology , Vascular Remodeling/physiology , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Female , Fluorescent Antibody Technique , Heart Failure/complications , Humans , Hypertension, Pulmonary/complications , Male , Mice , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Up-Regulation/physiology
7.
Blood ; 127(10): 1355-60, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26702059

ABSTRACT

Despite the increased risk of thrombosis in cancer patients compared with healthy individuals, mechanisms that regulate cancer-induced hypercoagulation are incompletely understood. The aim of this study was to investigate whether cell-specific hypoxia-inducible factor (HIF) 1α regulates cancer-associated hypercoagulation, using in vitro clotting assays and in vivo cancer models. In mouse lung and mammary tumor cells, hypoxia led to increases in cell adhesion, clotting, and fibrin deposition; these increases were eliminated in HIF1α null cells. Increased levels of HIF1α were also associated with increased tissue factor expression in human breast tumor samples. Conversely, deletion of endothelial (but not myeloid) cell-specific HIF1α doubled pulmonary fibrin deposition, and trebled thrombus formation compared with wildtype littermates in tumor-bearing mice. Our data suggest that tumor and endothelial cell-specific HIF1α may have opposing roles in cancer-associated coagulation and thrombosis. Off-target effects of manipulating the HIF1 axis in cancer patients should be carefully considered when managing thrombotic complications.


Subject(s)
Blood Coagulation , Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mammary Neoplasms, Experimental/metabolism , Thrombophilia/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Thrombophilia/genetics , Thrombophilia/pathology
8.
Am J Physiol Lung Cell Mol Physiol ; 312(4): L441-L451, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28130261

ABSTRACT

The prevailing morbidity and mortality in sepsis are largely due to multiple organ dysfunction (MOD), most commonly lung injury, as well as renal and cardiac dysfunction. Despite recent advances in defining many aspects of the pathogenesis of sepsis-related MOD, including acute respiratory distress syndrome (ARDS), there are currently no effective pharmacological or cell-based treatments for the disease. Human and animal studies have shown that pulmonary thrombosis is common in sepsis-induced ARDS, and preclinical studies have shown that anticoagulation may improve outcome following sepsis challenge. The potential beneficial effect of anticoagulation on outcome is unconvincing in clinical studies, however, and these discrepancies may arise from the multiple and sometimes opposing actions of thrombosis on the pulmonary endothelium following sepsis. It has been suggested, for example, that mild pulmonary thrombosis prevents escape of bacterial infection into the circulation, while severe thrombosis causes hypoxia and results in pulmonary endothelial damage. Evidence from both human and animal studies has demonstrated the key role of microvascular leakage in determining the outcome of sepsis. In this review, we describe thrombosis-dependent mechanisms that regulate pulmonary endothelial injury and repair following sepsis, including activation of the coagulation cascade by tissue factor and stimulation of vascular repair by hypoxia-inducible factors. Targeting such mechanisms through anticoagulant, anti-inflammatory, and reparative methods may represent a novel approach for the treatment of septic patients.


Subject(s)
Endothelium, Vascular/pathology , Lung Injury/etiology , Lung Injury/pathology , Sepsis/complications , Thrombosis/pathology , Wound Healing , Animals , Humans , Models, Biological
9.
Arterioscler Thromb Vasc Biol ; 36(2): 245-52, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26681755

ABSTRACT

Deep vein thrombosis and common complications, including pulmonary embolism and post-thrombotic syndrome, represent a major source of morbidity and mortality worldwide. Experimental models of venous thrombosis have provided considerable insight into the cellular and molecular mechanisms that regulate thrombus formation and subsequent resolution. Here, we critically appraise the ex vivo and in vivo techniques used to assess venous thrombosis in these models. Particular attention is paid to imaging modalities, including magnetic resonance imaging, micro-computed tomography, and high-frequency ultrasound that facilitate longitudinal assessment of thrombus size and composition.


Subject(s)
Veins , Venous Thrombosis/diagnosis , Animals , Biomarkers/metabolism , Blood Flow Velocity , Diagnostic Imaging/methods , Predictive Value of Tests , Prognosis , Regional Blood Flow , Veins/metabolism , Veins/pathology , Veins/physiopathology , Venous Thrombosis/metabolism , Venous Thrombosis/pathology , Venous Thrombosis/physiopathology
10.
Adv Exp Med Biol ; 967: 33-45, 2017.
Article in English | MEDLINE | ID: mdl-29047079

ABSTRACT

Pulmonary hypertension (PH) is a lung vascular disease with marked increases in pulmonary vascular resistance and pulmonary artery pressure (>25 mmHg at rest). In PH patients, increases in pulmonary vascular resistance lead to impaired cardiac output and reduced exercise tolerance. If untreated, PH progresses to right heart failure and premature lethality. The mechanisms that control the pathogenesis of PH are incompletely understood, but evidence from human and animal studies implicate nitrative stress in the development of PH. Increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) result in nitrative stress, which in turn induces posttranslational modification of key proteins important for maintaining pulmonary vascular homeostasis. This affects their functions and thereby contributes to the pathogenesis of PH. In this chapter, molecular mechanisms underlying nitrative stress-induced PH are reviewed, molecular sources of ROS and RNS are delineated, and evidence of nitrative stress in PH patients is described. A better understanding of such mechanisms could lead to the development of novel treatments for PH.


Subject(s)
Hypertension, Pulmonary/metabolism , Nitrates/metabolism , Oxidative Stress , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans , Hypertension, Pulmonary/physiopathology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Vascular Resistance
11.
Adv Exp Med Biol ; 906: 115-122, 2017.
Article in English | MEDLINE | ID: mdl-27638621

ABSTRACT

Venous thrombosis is a common complication in cancer patients, and some cancer chemotherapies are associated with an increased risk of venous thromboembolism. The regulatory mechanisms that control thrombus formation and subsequent resolution in patients with cancer, however, are incompletely understood, and novel treatments for cancer-associated thrombosis may arise from a better understanding of such mechanisms. In this chapter, pathways that regulate cancer-associated thrombus formation are outlined, and the effects of anti-angiogenic cancer chemotherapies on venous thrombus resolution are highlighted. Potentially pro-thrombotic effects of anti-angiogenic agents are important considerations when managing the complications of venous thrombosis in cancer patients.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Neoplasms/complications , Venous Thromboembolism/chemically induced , Venous Thrombosis/complications , Angiogenesis Inhibitors/administration & dosage , Disease Management , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Odds Ratio , Plasminogen Activator Inhibitor 1/blood , Risk , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/blood , Venous Thromboembolism/pathology , Venous Thrombosis/drug therapy , Venous Thrombosis/pathology
12.
FASEB J ; 29(3): 1102-12, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25422368

ABSTRACT

In mammals, hypoxia-triggered erythropoietin release increases red blood cell mass to meet tissue oxygen demands. Using male Wistar rats, we unmask a previously unrecognized regulatory pathway of erythropoiesis involving suppressor control by the NO metabolite and ubiquitous dietary component nitrate. We find that circulating hemoglobin levels are modulated by nitrate at concentrations achievable by dietary intervention under normoxic and hypoxic conditions; a moderate dose of nitrate administered via the drinking water (7 mg NaNO3/kg body weight/d) lowered hemoglobin concentration and hematocrit after 6 d compared with nonsupplemented/NaCl-supplemented controls. The underlying mechanism is suppression of hepatic erythropoietin expression associated with the downregulation of tissue hypoxia markers, suggesting increased pO2. At higher nitrate doses, however, a partial reversal of this effect occurred; this was accompanied by increased renal erythropoietin expression and stabilization of hypoxia-inducible factors, likely brought about by the relative anemia. Thus, hepatic and renal hypoxia-sensing pathways act in concert to modulate hemoglobin in response to nitrate, converging at an optimal minimal hemoglobin concentration appropriate to the environmental/physiologic situation. Suppression of hepatic erythropoietin expression by nitrate may thus act to decrease blood viscosity while matching oxygen supply to demand, whereas renal oxygen sensing could act as a brake, averting a potentially detrimental fall in hematocrit.


Subject(s)
Dietary Supplements , Erythropoiesis/drug effects , Erythropoietin/metabolism , Hemoglobins/metabolism , Hypoxia/metabolism , Nitrates/administration & dosage , Oxygen/metabolism , Animals , Epoetin Alfa , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Nitrates/pharmacology , Rats , Rats, Wistar , Recombinant Proteins/metabolism
13.
Arterioscler Thromb Vasc Biol ; 34(3): 565-70, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24436367

ABSTRACT

OBJECTIVE: Venous thromboembolism is a common complication in patients with cancer, resulting in significant morbidity and mortality. Clinical studies suggest that the incidence of venous thromboembolic events increased after treatment of these patients with antiangiogenic agents. Thrombi resolve through a process of remodeling, involving the formation of microvascular channels within the thrombus. Our aim was to determine whether inhibiting angiogenesis affects venous thrombus resolution. APPROACH AND RESULTS: Thrombus was induced in the inferior vena cava of mice. These mice were treated with axitinib (50 mg/kg per day), 2-methoxyestradiol (2ME, 150 mg/kg per day), or vehicle control. Thrombus size, recanalization, neovascularization, inflammatory cell content, and collagen content were assessed after axitinib (days 3, 10, 17) and 2ME (day 10 only) treatment (n=6/group). Axitinib treatment resulted in reduced thrombus resolution (P<0.002) and vein recanalization (P<0.001) compared with vehicle-treated controls. This was associated with inhibition of organization as seen through reduced thrombus neovascularization (P<0.0001) and collagen (P<0.0001) content, as well as reduced macrophage accumulation in the thrombus (P<0.001). Treatment with a second antiangiogenic agent, 2ME, mirrored these findings, with a similar order of magnitude of effect of treatment over vehicle control in all of the parameters measured, with the exception of neutrophil content, which was significantly reduced after 2ME treatment but not affected by axitinib. CONCLUSIONS: Antiangiogenic therapy (using axitinib and 2ME) inhibits the resolution of venous thrombi, which could lead to persistent venous obstruction and the possibility of thrombus extension. This potential prolongation of venous occlusion by antiangiogenic agents should therefore be taken into consideration in trials of these agents and when managing the complications of venous thromboembolic events in patients with cancer.


Subject(s)
Angiogenesis Inhibitors/toxicity , Blood Coagulation/drug effects , Estradiol/analogs & derivatives , Imidazoles/toxicity , Indazoles/toxicity , Venous Thrombosis/physiopathology , 2-Methoxyestradiol , Angiogenesis Inhibitors/pharmacology , Animals , Axitinib , Blood Coagulation/physiology , Capillary Permeability/drug effects , Collagen/analysis , Estradiol/pharmacology , Estradiol/toxicity , Imidazoles/pharmacology , Indazoles/pharmacology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Neutrophils/drug effects , Neutrophils/pathology , Thrombophilia/chemically induced , Vena Cava, Inferior , Venous Thrombosis/pathology
14.
Circulation ; 128(7): 729-736, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23820077

ABSTRACT

BACKGROUND: The magnetic resonance longitudinal relaxation time (T1) changes with thrombus age in humans. In this study, we investigate the possible mechanisms that give rise to the T1 signal in venous thrombi and whether changes in T1 relaxation time are informative of the susceptibility to lysis. METHODS AND RESULTS: Venous thrombosis was induced in the vena cava of BALB/C mice, and temporal changes in T1 relaxation time correlated with thrombus composition. The mean T1 relaxation time of thrombus was shortest at 7 days following thrombus induction and returned to that of blood as the thrombus resolved. T1 relaxation time was related to thrombus methemoglobin formation and further processing. Studies in inducible nitric oxide synthase (iNOS(-/-))-deficient mice revealed that inducible nitric oxide synthase mediates oxidation of erythrocyte lysis-derived iron to paramagnetic Fe3+, which causes thrombus T1 relaxation time shortening. Studies using chemokine receptor-2-deficient mice (Ccr2(-/-)) revealed that the return of the T1 signal to that of blood is regulated by removal of Fe3+ by macrophages that accumulate in the thrombus during its resolution. Quantification of T1 relaxation time was a good predictor of successful thrombolysis with a cutoff point of <747 ms having a sensitivity and specificity to predict successful lysis of 83% and 94%, respectively. CONCLUSIONS: The source of the T1 signal in the thrombus results from the oxidation of iron (released from the lysis of trapped erythrocytes in the thrombus) to its paramagnetic Fe3+ form. Quantification of T1 relaxation time appears to be a good predictor of the success of thrombolysis.


Subject(s)
Fibrinolysis/physiology , Iron/metabolism , Magnetic Resonance Imaging , Venous Thrombosis/pathology , Animals , Endothelium, Vascular/injuries , Erythrocytes/chemistry , Humans , Ligation , Macrophages/physiology , Male , Mass Spectrometry , Methemoglobin/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/physiology , Oxidation-Reduction , Receptors, CCR2/deficiency , Receptors, CCR2/physiology , Time Factors , Vena Cava, Inferior/pathology , Venous Thrombosis/etiology , Venous Thrombosis/metabolism
15.
medRxiv ; 2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37034726

ABSTRACT

Background: Patients with sepsis-induced acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) commonly suffer from severe pulmonary thrombosis, but clinical trials of anti-coagulant therapies in sepsis and ARDS patients have failed. ARDS patients with thrombocytopenia also exhibit increased mortality, and widespread pulmonary thrombosis is often seen in coronavirus disease 2019 (COVID-19) ARDS patients. Methods: Employing different amounts of microbeads to induce various levels of pulmonary thrombosis. Acute lung injury was induced by either lipopolysaccharide i.p. or cecal ligation and puncture. Endothelial cell (EC)-targeted nanoparticle coupled with CDH5 promoter was employed to delivery plasmid DNA expressing the CRISPR/Cas9 system for EC-specific gene knockout or expressing Alox15 for EC-specific overexpression. Additionally, thrombocytopenia was induced by genetic depletion of platelets using DTR Pf4Cre mice by breeding Pf4 Cre mice into the genetic background of DTR mice. Results: We show that while severe pulmonary thrombosis or thrombocytopenia augments sepsis-induced ALI, the induction of mild pulmonary thrombosis conversely reduces endothelial cell (EC) apoptosis, ALI, and mortality via sustained expression of endothelial arachidonate 15-lipoxygenase (Alox15). Endothelial Alox15 knockout via EC-targeted nanoparticle delivery of CRISPR/Cas9 plasmid DNA in adult mice abolished the protective impact of mild lung thrombosis. Conversely, overexpression of endothelial Alox15 inhibited the increases in ALI caused by severe pulmonary thrombosis. The clinical relevance of the findings was validated by the observation of reduced ALOX15-expressing ECs in lung autopsy samples of ARDS patients. Additionally, restoration of pulmonary thrombosis in thrombocytopenic mice also normalized endotoxemia-induced ALI. Conclusion: We have demonstrated that moderate levels of thrombosis protect against sepsis-induced inflammatory lung injury via endothelial Alox15. Overexpression of Alox5 inhibits severe pulmonary thrombosis-induced increase of ALI. Thus, activation of ALOX15 signaling represents a promising therapeutic strategy for treatment of ARDS, especially in sub-populations of patients with thrombocytopenia and/or severe pulmonary thrombosis.

16.
Sci Transl Med ; 15(709): eabm5755, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37585502

ABSTRACT

Aging is a major risk factor of high incidence and increased mortality of acute respiratory distress syndrome (ARDS). Here, we demonstrated that persistent lung injury and high mortality in aged mice after sepsis challenge were attributable to impaired endothelial regeneration and vascular repair. Genetic lineage tracing study showed that endothelial regeneration after sepsis-induced vascular injury was mediated by lung resident endothelial proliferation in young adult mice, whereas this intrinsic regenerative program was impaired in aged mice. Expression of forkhead box M1 (FoxM1), an important mediator of endothelial regeneration in young mice, was not induced in lungs of aged mice. Transgenic FOXM1 expression or in vivo endothelium-targeted nanoparticle delivery of the FOXM1 gene driven by an endothelial cell (EC)-specific promoter reactivated endothelial regeneration, normalized vascular repair and resolution of inflammation, and promoted survival in aged mice after sepsis challenge. In addition, treatment with the FDA-approved DNA demethylating agent decitabine was sufficient to reactivate FoxM1-dependent endothelial regeneration in aged mice, reverse aging-impaired resolution of inflammatory injury, and promote survival. Mechanistically, aging-induced Foxm1 promoter hypermethylation in mice, which could be inhibited by decitabine treatment, inhibited Foxm1 induction after sepsis challenge. In COVID-19 lung autopsy samples, FOXM1 was not induced in vascular ECs of elderly patients in their 80s, in contrast with middle-aged patients (aged 50 to 60 years). Thus, reactivation of FoxM1-mediated endothelial regeneration and vascular repair may represent a potential therapy for elderly patients with ARDS.


Subject(s)
COVID-19 , Forkhead Box Protein M1 , Lung Injury , Respiratory Distress Syndrome , Sepsis , Animals , Mice , Decitabine/pharmacology , Endothelium, Vascular/physiology , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Lung/metabolism , Lung Injury/genetics , Mice, Transgenic , Regeneration/physiology , Sepsis/metabolism
17.
Arterioscler Thromb Vasc Biol ; 31(3): 506-12, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21325673

ABSTRACT

Observational studies have shown that inflammatory cells accumulate within the thrombus and surrounding vein wall during the natural history of venous thrombosis. More recent studies have begun to unravel the mechanisms that regulate this interaction and have confirmed that thrombosis and inflammation are intimately linked. This review outlines our current knowledge of the complex relationship between inflammatory cell activity and venous thrombosis and highlights new areas of research in this field. A better understanding of this relationship could lead to the development of novel therapeutic targets that inhibit thrombus formation or promote its resolution.


Subject(s)
Blood Coagulation/immunology , Inflammation/immunology , Leukocytes/immunology , Venous Thrombosis/immunology , Animals , Endothelial Cells/immunology , Erythrocytes/immunology , Humans , Inflammation/blood , Inflammation Mediators/metabolism , Phagocytes/immunology , Signal Transduction , Venous Thrombosis/blood
18.
Cells ; 11(2)2022 01 06.
Article in English | MEDLINE | ID: mdl-35053299

ABSTRACT

Inflammatory lung injury is characterized by lung endothelial cell (LEC) death, alveolar epithelial cell (AEC) death, LEC-LEC junction weakening, and leukocyte infiltration, which together disrupt nutrient and oxygen transport. Subsequently, lung vascular repair is characterized by LEC and AEC regeneration and LEC-LEC junction re-annealing, which restores nutrient and oxygen delivery to the injured tissue. Pulmonary hypoxia is a characteristic feature of several inflammatory lung conditions, including acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and severe coronavirus disease 2019 (COVID-19). The vascular response to hypoxia is controlled primarily by the hypoxia-inducible transcription factors (HIFs) 1 and 2. These transcription factors control the expression of a wide variety of target genes, which in turn mediate key pathophysiological processes including cell survival, differentiation, migration, and proliferation. HIF signaling in pulmonary cell types such as LECs and AECs, as well as infiltrating leukocytes, tightly regulates inflammatory lung injury and repair, in a manner that is dependent upon HIF isoform, cell type, and injury stimulus. The aim of this review is to describe the HIF-dependent regulation of inflammatory lung injury and vascular repair. The review will also discuss potential areas for future study and highlight putative targets for inflammatory lung conditions such as ALI/ARDS and severe COVID-19. In the development of HIF-targeted therapies to reduce inflammatory lung injury and/or enhance pulmonary vascular repair, it will be vital to consider HIF isoform- and cell-specificity, off-target side-effects, and the timing and delivery strategy of the therapeutic intervention.


Subject(s)
Acute Lung Injury/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , COVID-19/metabolism , Hypoxia-Inducible Factor 1/metabolism , Lung/metabolism , Respiratory Distress Syndrome/metabolism , SARS-CoV-2/metabolism , Signal Transduction , Acute Lung Injury/pathology , COVID-19/pathology , Humans , Lung/pathology , Respiratory Distress Syndrome/pathology
19.
Biomolecules ; 12(10)2022 09 23.
Article in English | MEDLINE | ID: mdl-36291563

ABSTRACT

Cancer patients have a greater risk of thrombosis than individuals without cancer. Conversely, thrombosis is a diagnostic predictor of cancer, but the mechanisms by which thrombosis promotes tumor propagation are incompletely understood. Our previous studies showed that hypoxia-inducible factors (HIF) 1α and HIF2α are stabilized in myeloid cells of murine thrombi. We also previously showed that pulmonary thrombosis increases the levels of HIF1α and HIF2α in murine lungs, enhances the levels of tumorigenic factors in the circulation, and promotes pulmonary tumorigenesis. In this study, we aimed to investigate the regulation of thrombosis-induced tumorigenesis by myeloid cell-specific HIFs (i.e., HIF1 and HIF2 in neutrophils and macrophages). Our in vitro studies showed that multiple tumorigenic factors are upregulated in the secretome of hypoxic versus normoxic neutrophils and macrophages, which promotes lung cancer cell proliferation and migration in a myeloid-HIF-dependent manner. Next, we used a mouse model of pulmonary microvascular occlusion to study the impact of pulmonary thrombosis and myeloid HIFs on lung tumorigenesis. Experiments on mice lacking either HIF1α or HIF2α in myeloid cells demonstrated that loss of either factor eliminates the advantage given to pulmonary tumor formation by thrombotic insult. The myeloid HIF-dependent and tumorigenic impact of pulmonary thrombosis on tumor burden may be partly driven by paracrine thymidine phosphorylase (TP), given that TP levels were increased by hypoxia in neutrophil and macrophage supernates, and that plasma TP levels were positively correlated with multiple measures of tumor progression in wild type mice but not myeloid cell-specific HIF1α or HIF2α knockout mice. These data together demonstrate the importance of thrombotic insult in a model of pulmonary tumorigenesis and the essential role of myeloid HIFs in mediating tumorigenic success.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Thymidine Phosphorylase , Mice , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis , Mice, Knockout , Hypoxia/metabolism , Lung/metabolism
20.
Open Forum Infect Dis ; 9(5): ofac160, 2022 May.
Article in English | MEDLINE | ID: mdl-35493120

ABSTRACT

We present a novel case of recurrent brain abscesses found to be the result of a silent congenital right-to-left extracardiac shunt, a persistent left superior vena cava draining into the left atrium. The patient's brain abscess was evacuated surgically and treated with antibiotics, and his shunt was subsequently repaired. The case suggests that attention should be paid to evaluation for shunt physiology allowing for bypass of the pulmonary circulation in those with recurrent brain abscesses.

SELECTION OF CITATIONS
SEARCH DETAIL