Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 43(8): 1412-1428, 2023 08.
Article in English | MEDLINE | ID: mdl-37317853

ABSTRACT

BACKGROUND: During infectious diseases, proinflammatory cytokines transiently destabilize interactions between adjacent vascular endothelial cells (ECs) to facilitate the passage of immune molecules and cells into tissues. However, in the lung, the resulting vascular hyperpermeability can lead to organ dysfunction. Previous work identified the transcription factor ERG (erythroblast transformation-specific-related gene) as a master regulator of endothelial homeostasis. Here we investigate whether the sensitivity of pulmonary blood vessels to cytokine-induced destabilization is due to organotypic mechanisms affecting the ability of endothelial ERG to protect lung ECs from inflammatory injury. METHODS: Cytokine-dependent ubiquitination and proteasomal degradation of ERG were analyzed in cultured HUVECs (human umbilical vein ECs). Systemic administration of TNFα (tumor necrosis factor alpha) or the bacterial cell wall component lipopolysaccharide was used to cause a widespread inflammatory challenge in mice; ERG protein levels were assessed by immunoprecipitation, immunoblot, and immunofluorescence. Murine Erg deletion was genetically induced in ECs (Ergfl/fl;Cdh5[PAC]-CreERT2), and multiple organs were analyzed by histology, immunostaining, and electron microscopy. RESULTS: In vitro, TNFα promoted the ubiquitination and degradation of ERG in HUVECs, which was blocked by the proteasomal inhibitor MG132. In vivo, systemic administration of TNFα or lipopolysaccharide resulted in a rapid and substantial degradation of ERG within lung ECs but not ECs of the retina, heart, liver, or kidney. Pulmonary ERG was also downregulated in a murine model of influenza infection. Ergfl/fl;Cdh5(PAC)-CreERT2 mice spontaneously recapitulated aspects of inflammatory challenges, including lung-predominant vascular hyperpermeability, immune cell recruitment, and fibrosis. These phenotypes were associated with a lung-specific decrease in the expression of Tek-a gene target of ERG previously implicated in maintaining pulmonary vascular stability during inflammation. CONCLUSIONS: Collectively, our data highlight a unique role for ERG in pulmonary vascular function. We propose that cytokine-induced ERG degradation and subsequent transcriptional changes in lung ECs play critical roles in the destabilization of pulmonary blood vessels during infectious diseases.


Subject(s)
Communicable Diseases , Transcription Factors , Humans , Mice , Animals , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Lipopolysaccharides/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Cytokines/metabolism , Communicable Diseases/metabolism , Cells, Cultured , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
2.
PLoS Med ; 19(2): e1003911, 2022 02.
Article in English | MEDLINE | ID: mdl-35192610

ABSTRACT

BACKGROUND: There is limited evidence on the use of high-sensitivity C-reactive protein (hsCRP) as a biomarker for selecting patients for advanced cardiovascular (CV) therapies in the modern era. The prognostic value of mildly elevated hsCRP beyond troponin in a large real-world cohort of unselected patients presenting with suspected acute coronary syndrome (ACS) is unknown. We evaluated whether a mildly elevated hsCRP (up to 15 mg/L) was associated with mortality risk, beyond troponin level, in patients with suspected ACS. METHODS AND FINDINGS: We conducted a retrospective cohort study based on the National Institute for Health Research Health Informatics Collaborative data of 257,948 patients with suspected ACS who had a troponin measured at 5 cardiac centres in the United Kingdom between 2010 and 2017. Patients were divided into 4 hsCRP groups (<2, 2 to 4.9, 5 to 9.9, and 10 to 15 mg/L). The main outcome measure was mortality within 3 years of index presentation. The association between hsCRP levels and all-cause mortality was assessed using multivariable Cox regression analysis adjusted for age, sex, haemoglobin, white cell count (WCC), platelet count, creatinine, and troponin. Following the exclusion criteria, there were 102,337 patients included in the analysis (hsCRP <2 mg/L (n = 38,390), 2 to 4.9 mg/L (n = 27,397), 5 to 9.9 mg/L (n = 26,957), and 10 to 15 mg/L (n = 9,593)). On multivariable Cox regression analysis, there was a positive and graded relationship between hsCRP level and mortality at baseline, which remained at 3 years (hazard ratio (HR) (95% CI) of 1.32 (1.18 to 1.48) for those with hsCRP 2.0 to 4.9 mg/L and 1.40 (1.26 to 1.57) and 2.00 (1.75 to 2.28) for those with hsCRP 5 to 9.9 mg/L and 10 to 15 mg/L, respectively. This relationship was independent of troponin in all suspected ACS patients and was further verified in those who were confirmed to have an ACS diagnosis by clinical coding. The main limitation of our study is that we did not have data on underlying cause of death; however, the exclusion of those with abnormal WCC or hsCRP levels >15 mg/L makes it unlikely that sepsis was a major contributor. CONCLUSIONS: These multicentre, real-world data from a large cohort of patients with suspected ACS suggest that mildly elevated hsCRP (up to 15 mg/L) may be a clinically meaningful prognostic marker beyond troponin and point to its potential utility in selecting patients for novel treatments targeting inflammation. TRIAL REGISTRATION: ClinicalTrials.gov - NCT03507309.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/mortality , C-Reactive Protein/metabolism , Acute Coronary Syndrome/diagnosis , Aged , Aged, 80 and over , Biomarkers/blood , Cohort Studies , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Mortality/trends , Predictive Value of Tests , Retrospective Studies , Risk Factors , United Kingdom/epidemiology
3.
Circ Res ; 127(7): 928-944, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32611235

ABSTRACT

RATIONALE: The efficient resolution of tissue hemorrhage is an important homeostatic function. In human macrophages in vitro, heme activates an AMPK (AMP-activated protein kinase)/ATF1 (activating transcription factor-1) pathway that directs Mhem macrophages through coregulation of HO-1 (heme oxygenase-1; HMOX1) and lipid homeostasis genes. OBJECTIVE: We asked whether this pathway had an in vivo role in mice. METHODS AND RESULTS: Perifemoral hematomas were used as a model of hematoma resolution. In mouse bone marrow-derived macrophages, heme induced HO-1, lipid regulatory genes including LXR (lipid X receptor), the growth factor IGF1 (insulin-like growth factor-1), and the splenic red pulp macrophage gene Spic. This response was lost in bone marrow-derived macrophages from mice deficient in AMPK (Prkab1-/-) or ATF1 (Atf1-/-). In vivo, femoral hematomas resolved completely between days 8 and 9 in littermate control mice (n=12), but were still present at day 9 in mice deficient in either AMPK (Prkab1-/-) or ATF1 (Atf1-/-; n=6 each). Residual hematomas were accompanied by increased macrophage infiltration, inflammatory activation and oxidative stress. We also found that fluorescent lipids and a fluorescent iron-analog were trafficked to lipid-laden and iron-laden macrophages respectively. Moreover erythrocyte iron and lipid abnormally colocalized in the same macrophages in Atf1-/- mice. Therefore, iron-lipid separation was Atf1-dependent. CONCLUSIONS: Taken together, these data demonstrate that both AMPK and ATF1 are required for normal hematoma resolution. Graphic Abstract: An online graphic abstract is available for this article.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Activating Transcription Factor 1/metabolism , Hematoma/metabolism , Macrophages/metabolism , AMP-Activated Protein Kinases/genetics , Activating Transcription Factor 1/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Erythrocytes/metabolism , Female , Hematoma/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Iron/metabolism , Lipid Metabolism , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Time Factors
4.
Clin Exp Rheumatol ; 37 Suppl 121(6): 48-51, 2019.
Article in English | MEDLINE | ID: mdl-31172922

ABSTRACT

OBJECTIVES: Behçet's syndrome (BS) is a rare multi-system inflammatory disorder. Clinical phenotypic variance across geographical regions is recognised but UK BS patients' variance by age groups and gender has not been studied. This study compares the clinical features of adult and juvenile onset Behçet's Syndrome (JBS) in a UK population. METHODS: Two clinical databases of BS patients were compared. The JBS database was collected at the Great Ormond Street Hospital for Children, London (n=46). The adult database was collected at the Hammersmith Hospital, London (n=560). RESULTS: Oro-genital aphthosis had high prevalence in both the JBS and the adult cohort (oral: 97.8% vs. 96.6%, genital: 73.9% vs. 75.7%). The JBS cohort was more likely to have gastrointestinal involvement (21.7% vs. 4.5%, p<0.001) and arthritis (21.7% vs. 9.6%, p=0.021) compared to adults. The JBS cohort was less likely to have eye involvement (4.3% vs. 37%, p<0.001), skin (21.7% vs. 55.4%, p<0.001) and vascular involvement (6.5% vs. 17.5% p=0.063). JBS females had a higher rate of genital aphthosis than JBS males (87.5% vs. 59.1%, p=0.044). Adult females had higher rates of genital (85.2% vs. 64.5%, p<0.001) and oral (99.0% vs. 93.8%, p=0.001) aphthosis than adult males. Adult males were more likely to have ophthalmological (44.9% vs. 30.3%, p<0.001) and vascular (23.0% vs. 12.8%, p=0.002) manifestations than adult females. CONCLUSIONS: UK JBS patients displayed less ocular and skin manifestations compared to the adult BS patients. This information will aid clinicians in diagnosing BS in UK adult and paediatric populations.


Subject(s)
Behcet Syndrome , Adult , Age Factors , Age of Onset , Behcet Syndrome/pathology , Behcet Syndrome/physiopathology , Child , Cohort Studies , Female , Genital Diseases, Female/diagnosis , Genital Diseases, Female/etiology , Genital Diseases, Male/diagnosis , Genital Diseases, Male/etiology , Humans , Male , Retinal Vasculitis/diagnosis , Retinal Vasculitis/etiology , Sex Factors , Stomatitis, Aphthous/diagnosis , Stomatitis, Aphthous/etiology , Symptom Assessment , United Kingdom
5.
Blood ; 137(10): 1280-1281, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33704393
6.
Arterioscler Thromb Vasc Biol ; 37(1): 130-143, 2017 01.
Article in English | MEDLINE | ID: mdl-27834691

ABSTRACT

OBJECTIVE: Atherosclerosis is initiated at branches and bends of arteries exposed to disturbed blood flow that generates low shear stress. This mechanical environment promotes lesions by inducing endothelial cell (EC) apoptosis and dysfunction via mechanisms that are incompletely understood. Although transcriptome-based studies have identified multiple shear-responsive genes, most of them have an unknown function. To address this, we investigated whether zebrafish embryos can be used for functional screening of mechanosensitive genes that regulate EC apoptosis in mammalian arteries. APPROACH AND RESULTS: First, we demonstrated that flow regulates EC apoptosis in developing zebrafish vasculature. Specifically, suppression of blood flow in zebrafish embryos (by targeting cardiac troponin) enhanced that rate of EC apoptosis (≈10%) compared with controls exposed to flow (≈1%). A panel of candidate regulators of apoptosis were identified by transcriptome profiling of ECs from high and low shear stress regions of the porcine aorta. Genes that displayed the greatest differential expression and possessed 1 to 2 zebrafish orthologues were screened for the regulation of apoptosis in zebrafish vasculature exposed to flow or no-flow conditions using a knockdown approach. A phenotypic change was observed in 4 genes; p53-related protein (PERP) and programmed cell death 2-like protein functioned as positive regulators of apoptosis, whereas angiopoietin-like 4 and cadherin 13 were negative regulators. The regulation of perp, cdh13, angptl4, and pdcd2l by shear stress and the effects of perp and cdh13 on EC apoptosis were confirmed by studies of cultured EC exposed to flow. CONCLUSIONS: We conclude that a zebrafish model of flow manipulation coupled to gene knockdown can be used for functional screening of mechanosensitive genes in vascular ECs, thus providing potential therapeutic targets to prevent or treat endothelial injury at atheroprone sites.


Subject(s)
Apoptosis , Atherosclerosis/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Mechanotransduction, Cellular/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Embryo, Nonmammalian/blood supply , Endothelial Cells/pathology , Female , Gene Expression Profiling/methods , Gene Knockdown Techniques , Gene Regulatory Networks , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Mice , Phenotype , RNA Interference , Regional Blood Flow , Stress, Mechanical , Swine , Transcriptome , Transfection , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Mol Imaging ; 16: 1536012117723788, 2017.
Article in English | MEDLINE | ID: mdl-28877655

ABSTRACT

The identification of vulnerable coronary artery atherosclerotic plaques offers the prospect of either localized or systematic therapeutic targeting in order to prevent myocardial infarction. Molecular imaging of atherosclerosis adds to morphological imaging by focusing on the immunobiology hidden in and behind the endothelium and therefore may be able to improve the identification of prospective culprit lesions. Our focus has been on identifying arterial accumulation of oxidized low-density lipoprotein (oxLDL) by exploiting advances in knowledge of vascular pathobiology. Here, we reflect on our work developing near-infrared fluorescence imaging of oxLDL using LO1, a monoclonal autoantibody generated in our laboratory. We detail progress to date and discuss our vision on taking the work through the early translational pipeline toward a multitargeted approach in imaging rupture-prone atherosclerotic plaques. Ultimately, molecular imaging of coronary arteries should be able to assess the regional risk that is specific to a lesion, which can then be used in concert with global risk factors to personalize the therapeutic strategy for patients in a way that goes beyond generalized population-based therapies.


Subject(s)
Atherosclerosis/metabolism , Lipoproteins, LDL/metabolism , Molecular Imaging/methods , Autoantibodies/metabolism , Humans , Spectroscopy, Near-Infrared
8.
Blood ; 124(24): 3646-55, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25293769

ABSTRACT

Tissue factor (TF) (CD142) is a 47 kDa transmembrane cell surface glycoprotein that triggers the extrinsic coagulation cascade and links thrombosis with inflammation. Although macrophage TF expression is known to be regulated at the RNA level, very little is known about the mechanisms involved. Poly(adenosine 5'-diphosphate [ADP]-ribose)-polymerase (PARP)-14 belongs to a family of intracellular proteins that generate ADP-ribose posttranslational adducts. Functional screening of PARP-14-deficient macrophages mice revealed that PARP-14 deficiency leads to increased TF expression and functional activity in macrophages after challenge with bacterial lipopolysaccharide. This was related to an increase in TF messenger RNA (mRNA) stability. Ribonucleoprotein complex immunoprecipitation and biotinylated RNA pull-down assays demonstrated that PARP-14 forms a complex with the mRNA-destabilizing protein tristetraprolin (TTP) and a conserved adenylate-uridylate-rich element in the TF mRNA 3' untranslated region. TF mRNA regulation by PARP-14 was selective, as tumor necrosis factor (TNF)α mRNA, which is also regulated by TTP, was not altered in PARP-14 deficient macrophages. Consistent with the in vitro data, TF expression and TF activity, but not TNFα expression, were increased in Parp14(-/-) mice in vivo. Our study provides a novel mechanism for the posttranscriptional regulation of TF expression, indicating that this is selectively regulated by PARP-14.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Thromboplastin/biosynthesis , Tristetraprolin/metabolism , 3' Untranslated Regions/physiology , Animals , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , RNA Stability/drug effects , RNA Stability/physiology , Thromboplastin/genetics , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
9.
J Immunol ; 192(9): 4316-27, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24670799

ABSTRACT

Endothelial injury and dysfunction precede accelerated arterial disease in allograft vasculopathy and systemic autoimmune diseases and involve pathogenic Abs and complement. Recent reports suggest that switching to rapamycin from calcineurin antagonists reduces posttransplant vasculopathy and prolongs survival following cardiac transplantion. The majority of these patients also receive statin therapy. We examined potential mechanisms underlying this protective response in human endothelial cells and identified synergy between rapamycin and atorvastatin. Mechanistically, atorvastatin and rapamycin activated a protein kinase Cα, AMP-activated kinase, and CREB-dependent vasculoprotective pathway, which induced decay-accelerating factor (DAF) promoter activity via binding to the cAMP response element, mutation of which attenuated promoter activity. This response significantly increased endothelial cell surface DAF and enhanced protection against complement-mediated injury. Synergy with rapamycin was reproduced by simvastatin, whereas combining atorvastatin with cyclosporine or mycophenolate in place of rapamycin was ineffective. Importantly, synergy was reproduced in vivo, in which only atorvastatin and rapamycin therapy in combination was sufficient to induce DAF on murine aortic endothelium. We believe this pathway represents an important therapeutically inducible vasculoprotective mechanism for diseases mediated by pathogenic Abs and complement, including posttransplant vasculopathy and systemic lupus erythematosus. Although our study focuses on the vascular endothelium, the findings are likely to be broadly applicable, given the diverse cellular expression of DAF.


Subject(s)
Cytoprotection/drug effects , Endothelium, Vascular/drug effects , Heptanoic Acids/administration & dosage , Pyrroles/administration & dosage , Signal Transduction/drug effects , Sirolimus/administration & dosage , AMP-Activated Protein Kinases/metabolism , Animals , Atorvastatin , CD55 Antigens/metabolism , Complement Activation/drug effects , Complement Activation/physiology , Complement System Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoprotection/physiology , Drug Synergism , Endothelium, Vascular/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Immunosuppressive Agents/administration & dosage , Mice , Protein Kinase C/metabolism , Signal Transduction/physiology
10.
Circ Res ; 112(12): 1583-91, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23564640

ABSTRACT

RATIONALE: Hypoxia followed by reoxygenation promotes inflammation by activating nuclear factor κB transcription factors in endothelial cells (ECs). This process involves modification of the signaling intermediary tumor necrosis factor receptor-associated factor 6 with polyubiquitin chains. Thus, cellular mechanisms that suppress tumor necrosis factor receptor-associated factor 6 ubiquitination are potential therapeutic targets to reduce inflammation in hypoxic tissues. OBJECTIVE: In this study, we tested the hypothesis that endothelial activation in response to hypoxia-reoxygenation can be influenced by Cezanne, a deubiquitinating enzyme that cleaves ubiquitin from specific modified proteins. METHODS AND RESULTS: Studies of cultured ECs demonstrated that hypoxia (1% oxygen) induced Cezanne via p38 mitogen-activated protein kinase-dependent transcriptional and post-transcriptional mechanisms. Hypoxia-reoxygenation had minimal effects on proinflammatory signaling in unmanipulated ECs but significantly enhanced Lys63 polyubiquitination of tumor necrosis factor receptor-associated factor 6, activation of nuclear factor κB, and expression of inflammatory genes after silencing of Cezanne. Thus, although hypoxia primed cells for inflammatory activation, it simultaneously induced Cezanne, which impeded signaling to nuclear factor κB by suppressing tumor necrosis factor receptor-associated factor 6 ubiquitination. Similarly, ischemia induced Cezanne in the murine kidney in vascular ECs, glomerular ECs, podocytes, and epithelial cells, and genetic deletion of Cezanne enhanced renal inflammation and injury in murine kidneys exposed to ischemia followed by reperfusion. CONCLUSIONS: We conclude that inflammatory responses to ischemia are controlled by a balance between ubiquitination and deubiquitination, and that Cezanne is a key regulator of this process. Our observations have important implications for therapeutic targeting of inflammation and injury during ischemia-reperfusion.


Subject(s)
Endopeptidases/metabolism , Endothelial Cells/enzymology , Inflammation/prevention & control , Kidney/blood supply , Reperfusion Injury/enzymology , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Endopeptidases/deficiency , Endopeptidases/genetics , Endothelial Cells/immunology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Oxygen/metabolism , RNA Interference , Rats , Rats, Inbred F344 , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , Time Factors , Transcription, Genetic , Transfection , Ubiquitination , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Arterioscler Thromb Vasc Biol ; 34(5): 985-95, 2014 May.
Article in English | MEDLINE | ID: mdl-24651677

ABSTRACT

OBJECTIVE: Although atherosclerosis is associated with systemic risk factors such as age, high cholesterol, and obesity, plaque formation occurs predominately at branches and bends that are exposed to disturbed patterns of blood flow. The molecular mechanisms that link disturbed flow-generated mechanical forces with arterial injury are uncertain. To illuminate them, we investigated the effects of flow on endothelial cell (EC) senescence. APPROACH AND RESULTS: LDLR(-/-) (low-density lipoprotein receptor(-/-)) mice were exposed to a high-fat diet for 2 to 12 weeks (or to a normal chow diet as a control) before the assessment of cellular senescence in aortic ECs. En face staining revealed that senescence-associated ß-galactosidase activity and p53 expression were elevated in ECs at sites of disturbed flow in response to a high-fat diet. By contrast, ECs exposed to undisturbed flow did not express senescence-associated ß-galactosidase or p53. Studies of aortae from healthy pigs (aged 6 months) also revealed enhanced senescence-associated ß-galactosidase staining at sites of disturbed flow. These data suggest that senescent ECs accumulate at disturbed flow sites during atherogenesis. We used in vitro flow systems to examine whether a causal relationship exists between flow and EC senescence. Exposure of cultured ECs to flow (using either an orbital shaker or a syringe-pump flow bioreactor) revealed that disturbed flow promoted EC senescence compared with static conditions, whereas undisturbed flow reduced senescence. Gene silencing studies demonstrated that disturbed flow induced EC senescence via a p53-p21 signaling pathway. Disturbed flow-induced senescent ECs exhibited reduced migration compared with nonsenescent ECs in a scratch wound closure assay, and thus may be defective for arterial repair. However, pharmacological activation of sirtuin 1 (using resveratrol or SRT1720) protected ECs from disturbed flow-induced senescence. CONCLUSIONS: Disturbed flow promotes endothelial senescence via a p53-p21-dependent pathway which can be inhibited by activation of sirtuin 1. These observations support the principle that pharmacological activation of sirtuin 1 may promote cardiovascular health by suppressing EC senescence at atheroprone sites.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cellular Senescence , Endothelial Cells/metabolism , Mechanotransduction, Cellular , Tumor Suppressor Protein p53/metabolism , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Bioreactors , Cell Movement , Cells, Cultured , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Enzyme Activators/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mechanotransduction, Cellular/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , Receptors, LDL/deficiency , Receptors, LDL/genetics , Regional Blood Flow , Sirtuin 1/metabolism , Stress, Mechanical , Swine , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Wound Healing
12.
Blood ; 119(3): 894-903, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22117042

ABSTRACT

The endothelial ETS transcription factor Erg plays an important role in homeostasis and angiogenesis by regulating many endothelial functions including survival and junction stability. Here we show that Erg regulates endothelial cell (EC) migration. Transcriptome profiling of Erg-deficient ECs identified ∼ 80 genes involved in cell migration as candidate Erg targets, including many regulators of Rho- GTPases. Inhibition of Erg expression in HUVECs resulted in decreased migration in vitro, while Erg overexpression using adenovirus caused increased migration. Live-cell imaging of Erg-deficient HUVECs showed a reduction in lamellipodia, in line with decreased motility. Both actin and tubulin cytoskeletons were disrupted in Erg-deficient ECs, with a dramatic increase in tubulin acetylation. Among the most significant microarray hits was the cytosolic histone deacetylase 6 (HDAC6), a regulator of cell migration. Chromatin immunoprecipitation (ChIP) and transactivation studies demonstrated that Erg regulates HDAC6 expression. Rescue experiments confirmed that HDAC6 mediates the Erg-dependent regulation of tubulin acetylation and actin localization. In vivo, inhibition of Erg expression in angiogenic ECs resulted in decreased HDAC6 expression with increased tubulin acetylation. Thus, we have identified a novel function for the transcription factor Erg in regulating HDAC6 and multiple pathways essential for EC migration and angiogenesis.


Subject(s)
Biomarkers/metabolism , Cell Movement , Endothelium, Vascular/metabolism , Gene Expression Regulation , Histone Deacetylases/genetics , Neovascularization, Physiologic , Signal Transduction , Trans-Activators/metabolism , Acetylation , Actins/metabolism , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Endothelium, Vascular/cytology , Gene Expression Profiling , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcriptional Regulator ERG , Umbilical Veins/cytology , Umbilical Veins/metabolism
13.
Cell Commun Signal ; 12: 12, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24593809

ABSTRACT

BACKGROUND: Endothelial junctions control functions such as permeability, angiogenesis and contact inhibition. VE-Cadherin (VECad) is essential for the maintenance of intercellular contacts. In confluent endothelial monolayers, N-Cadherin (NCad) is mostly expressed on the apical and basal membrane, but in the absence of VECad it localizes at junctions. Both cadherins are required for vascular development. The intercellular adhesion molecule (ICAM)-2, also localized at endothelial junctions, is involved in leukocyte recruitment and angiogenesis. RESULTS: In human umbilical vein endothelial cells (HUVEC), both VECad and NCad were found at nascent cell contacts of sub-confluent monolayers, but only VECad localized at the mature junctions of confluent monolayers. Inhibition of ICAM-2 expression by siRNA caused the appearance of small gaps at the junctions and a decrease in NCad junctional staining in sub-confluent monolayers. Endothelioma lines derived from WT or ICAM-2-deficient mice (IC2neg) lacked VECad and failed to form junctions, with loss of contact inhibition. Re-expression of full-length ICAM-2 (IC2 FL) in IC2neg cells restored contact inhibition through recruitment of NCad at the junctions. Mutant ICAM-2 lacking the binding site for ERM proteins (IC2 ΔERM) or the cytoplasmic tail (IC2 ΔTAIL) failed to restore junctions. ICAM-2-dependent Rac-1 activation was also decreased in these mutant cell lines. Barrier function, measured in vitro via transendothelial electrical resistance, was decreased in IC2neg cells, both in resting conditions and after thrombin stimulation. This was dependent on ICAM-2 signalling to the small GTPase Rac-1, since transendothelial electrical resistance of IC2neg cells was restored by constitutively active Rac-1. In vivo, thrombin-induced extravasation of FITC-labeled albumin measured by intravital fluorescence microscopy in the mouse cremaster muscle showed that permeability was increased in ICAM-2-deficient mice compared to controls. CONCLUSIONS: These results indicate that ICAM-2 regulates endothelial barrier function and permeability through a pathway involving N-Cadherin, ERMs and Rac-1.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Cell Adhesion Molecules/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antigens, CD/genetics , Binding Sites , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cytoskeletal Proteins/metabolism , Gap Junctions/metabolism , Humans , Membrane Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Protein Binding , Protein Transport , Signal Transduction
14.
Circ Res ; 110(1): 20-33, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22052915

ABSTRACT

RATIONALE: Intraplaque hemorrhage (IPH) drives atherosclerosis through the dual metabolic stresses of cholesterol-enriched erythrocyte membranes and pro-oxidant heme/iron. When clearing tissue hemorrhage, macrophages are typically seen storing either iron or lipid. We have recently defined hemorrhage-associated macrophages (HA-mac) as a plaque macrophage population that responds adaptively to IPH. OBJECTIVE: This study aimed to define the key transcription factor(s) involved in HO-1 induction by heme. METHODS AND RESULTS: To address this question, we used microarray analysis and transfection with siRNA and plasmids. To maintain physiological relevance, we focused on human blood-derived monocytes. We found that heme stimulates monocytes through induction of activating transcription factor 1 (ATF-1). ATF-1 coinduces heme oxygenase-1 (HO-1) and Liver X receptor beta (LXR-ß). Heme-induced HO-1 and LXR-ß were suppressed by knockdown of ATF-1, and HO-1 and LXR-ß were induced by ATF-1 transfection. ATF-1 required phosphorylation for full functional activity. Expression of LXR-ß in turn led to induction of other genes central to cholesterol efflux, such as LXR-α and ABCA1. This heme-directed state was distinct from known macrophage states (M1, M2, Mox) and, following the same format, we have designated them Mhem. CONCLUSIONS: These results show that ATF-1 mediates HO-1 induction by heme and drives macrophage adaptation to intraplaque hemorrhage. Our definition of an ATF-1-mediated pathway for linked protection from foam cell formation and oxidant stress may have therapeutic potential.


Subject(s)
Activating Transcription Factor 1/metabolism , Foam Cells/pathology , Hemorrhage/pathology , Iron/metabolism , Macrophages/metabolism , Macrophages/pathology , Plaque, Atherosclerotic/pathology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Cell Communication , Cells, Cultured , Heme/pharmacology , Heme Oxygenase-1/metabolism , Humans , Lipid Metabolism/physiology , Liver X Receptors , Macrophages/drug effects , Orphan Nuclear Receptors/metabolism , Oxidative Stress/physiology , Plaque, Atherosclerotic/physiopathology , Plaque, Atherosclerotic/prevention & control , Signal Transduction/physiology
15.
Arterioscler Thromb Vasc Biol ; 33(11): 2470-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24051143

ABSTRACT

OBJECTIVE: Intraplaque hemorrhage (IPH) is an important driver of the progression of atherosclerotic plaques. Recently, we characterized Mhem as a novel macrophage phenotype that limits the atherogenicity of IPH. Mhem are directed by activating transcription factor 1 (ATF1), which is activated by phosphorylation. A better understanding of the counteratherogenic ATF1-Mhem pathway may facilitate antiatherosclerotic therapies. APPROACH AND RESULTS: We tested the hypothesis that heme in pathologically relevant concentrations activates the ATF1-Mhem pathway via 5'-AMP-activated protein kinase (AMPK) in primary human monocyte-derived macrophages and mouse bone marrow macrophages. We found that heme (10 µmol/L) activates AMPK, and downstream ATF1-mediated coinduction of heme oxygenase and liver X receptor that characterize Mhem. Heme increased macrophage phospho-AMPK, phospho-ATF1, and its target genes, and these effects were inhibited by the AMPK antagonist dorsomorphin, or by AMPK-knockdown with small inhibitory ribonucleic acid. The AMPK-activating oral hypoglycemic agent metformin also induced and phosphorylated ATF1 at a clinically relevant concentration (10 µmol/L). Functional effects of heme and metformin were inhibited by AMPK-knockdown and included suppression of macrophage oxidative stress; increased cholesterol export; protection from foam-cell formation; and suppression of macrophage inflammatory activation (human leukocyte antigen type DR expression). CONCLUSIONS: Our data indicate that heme activates the ATF1 pathway in human macrophages via AMPK, and that a similar response occurs after treatment of cells with metformin. Our results suggest an in vitro mechanism that may explain the clinical evidence that metformin has vascular protective effects beyond its role in treating hyperglycemia.


Subject(s)
Activating Transcription Factor 1/metabolism , Atherosclerosis/metabolism , Heme/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Metformin/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Activating Transcription Factor 1/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Hypoglycemic Agents/pharmacology , Liver X Receptors , Macrophages/cytology , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Primary Cell Culture , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Clin Exp Rheumatol ; 32(4 Suppl 84): S99-102, 2014.
Article in English | MEDLINE | ID: mdl-25268665

ABSTRACT

OBJECTIVES: Vascular disease is a serious complication of Behçet's syndrome (BS), occurring in up to 20% of subjects. Superficial thrombophlebitis, deep vein thrombosis, and arterial aneurysm formation are the most common manifestations. Venous thrombosis is thought to result from vessel wall inflammation. This work investigated the potential usefulness of high resolution magnetic resonance imaging (MRI) to identify inflammation in the venous walls in BS subjects. METHODS: Seven healthy control (HC) subjects and five BS subjects were scanned with 3T MRI (Siemens Skyra). A standard MRI sequence was adapted for use in the venous system. Metronome guided breathing generated a regular respiratory variation of venous blood velocity. The vein wall imaging was triggered at an appropriate delay after the metronome. The popliteal vein was imaged. Vein wall images were ranked based on wall thickness and signal enhancement by two blinded, experienced observers. RESULTS: Popliteal vein rank scores were found to be significantly increased in BS versus HC subjects by the first observer (p(Observer 1)=0.025, p(Observer2)=0.07) and also averaging both observers (p=0.05). The repeated images of each subject gave a degree of variability in results, potentially from drifting response to metronome guidance over the 10 minute scan. CONCLUSIONS: MR imaging can detect increased vein wall thickness in BS subjects compared to healthy controls. Variable response to the metronome-guided breathing requires further development.


Subject(s)
Behcet Syndrome/pathology , Magnetic Resonance Angiography/methods , Popliteal Vein/pathology , Venous Thrombosis/pathology , Adult , Feasibility Studies , Female , Humans , Magnetic Resonance Angiography/statistics & numerical data , Male , Middle Aged , Observer Variation , Vascular Diseases/pathology , Young Adult
17.
RSC Chem Biol ; 5(7): 640-651, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38966672

ABSTRACT

The post-translational modification (PTM) ADP-ribosylation plays an important role in cell signalling and regulating protein function and has been implicated in the development of multiple diseases, including breast and ovarian cancers. Studying the underlying mechanisms through which this PTM contributes towards disease development, however, has been hampered by the lack of appropriate tools for reliable identification of physiologically relevant ADP-ribosylated proteins in a live-cell environment. Herein, we explore the application of an alkyne-tagged proprobe, 6Yn-ProTide-Ad (6Yn-Pro) as a chemical tool for the identification of intracellular ADP-ribosylated proteins through metabolic labelling. We applied targeted metabolomics and chemical proteomics in HEK293T cells treated with 6Yn-Pro to demonstrate intracellular metabolic conversion of the probe into ADP-ribosylation cofactor 6Yn-NAD+, and subsequent labelling and enrichment of PARP1 and multiple known ADP-ribosylated proteins in cells under hydrogen peroxide-induced stress. We anticipate that the approach and methodology described here will be useful for future identification of novel intracellular ADP-ribosylated proteins.

18.
J Biol Chem ; 287(15): 12331-42, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22337883

ABSTRACT

The interaction of transcription factors with specific DNA sequences is critical for activation of gene expression programs. In endothelial cells (EC), the transcription factor NF-κB is important in the switch from quiescence to activation, and is tightly controlled to avoid excessive inflammation and organ damage. Here we describe a novel mechanism that controls the activation of NF-κB in EC. The transcription factor Erg, the most highly expressed ETS member in resting EC, controls quiescence by repressing proinflammatory gene expression. Focusing on intercellular adhesion molecule 1(ICAM)-1 as a model, we identify two ETS binding sites (EBS -118 and -181) within the ICAM-1 promoter required for Erg-mediated repression. We show that Erg binds to both EBS -118 and EBS -181, the latter located within the NF-κB binding site. Interestingly, inhibition of Erg expression in quiescent EC results in increased NF-κB-dependent ICAM-1 expression, indicating that Erg represses basal NF-κB activity. Erg prevents NF-κB p65 from binding to the ICAM-1 promoter, suggesting a direct mechanism of interference. Gene set enrichment analysis of transcriptome profiles of Erg and NF-κB-dependent genes, together with chromatin immunoprecipitation (ChIP) studies, reveals that this mechanism is common to other proinflammatory genes, including cIAP-2 and IL-8. These results identify a role for Erg as a gatekeeper controlling vascular inflammation, thus providing an important barrier to protect against inappropriate endothelial activation.


Subject(s)
Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/physiology , Trans-Activators/physiology , Transcription Factor RelA/metabolism , Binding Sites , Binding, Competitive , Cells, Cultured , DNA/chemistry , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Genes, Reporter , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Promoter Regions, Genetic , Protein Binding , Resting Phase, Cell Cycle , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcription Initiation Site , Transcription, Genetic , Transcriptional Regulator ERG
19.
Microcirculation ; 20(3): 203-16, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23121167

ABSTRACT

Atherosclerosis is a chronic inflammatory disease of the medium and large arteries driven in large part by the accumulation of oxidized low-density lipoproteins and other debris at sites rendered susceptible because of the geometry of the arterial tree. As lesions develop, they acquire a pathologic microcirculation that perpetuates lesion progression, both by providing a means for further monocyte and T-lymphocyte recruitment into the arterial wall and by the physical and chemical stresses caused by micro-hemorrhage. This review summarizes work performed in our department investigating the roles of signaling pathways, alone and in combination, that lead to specific programs of gene expression in the atherosclerotic environment. Focusing particularly on cytoprotective responses that might be enhanced therapeutically, the work has encompassed the anti-inflammatory effects of arterial laminar shear stress, mechanisms of induction of membrane inhibitors that prevent complement-mediated injury, homeostatic macrophage responses to hemorrhage, and the transcriptional mechanisms that control the stability, survival, and quiescence of endothelial monolayers. Lastly, while the field has been dominated by investigation into the mechanisms of DNA transcription, we consider the importance of parallel post-transcriptional regulatory mechanisms for fine-tuning functional gene expression repertoires.


Subject(s)
Cell Communication , Endothelial Cells/metabolism , Gene Expression Regulation , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Signal Transduction , Animals , Endothelial Cells/pathology , Humans , Macrophages/pathology , Plaque, Atherosclerotic/pathology
20.
Am J Pathol ; 181(1): 347-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22658487

ABSTRACT

IgG may accelerate atherosclerosis via ligation of proinflammatory Fcγ receptors; however, IgM is unable to ligate FcγR and is often considered vasculoprotective. IgM aggravates ischemia-reperfusion injury, and solid-phase deposits of pure IgM, as seen with IgM-secreting neoplasms, are well known clinically to provoke vascular inflammation. We therefore examined the molecular mechanisms by which immunoglobulins can aggravate vascular inflammation, such as in atherosclerosis. We compared the ability of fluid- and solid-phase immunoglobulins to activate macrophages. Solid-phase immunoglobulins initiated prothrombotic and proinflammatory functions in human macrophages, including NF-κB p65 activation, H(2)O(2) secretion, macrophage-induced apoptosis, and tissue factor expression. Responses to solid-phase IgG (but not to IgM) were blocked by neutralizing antibodies to CD16 (FcγRIII), consistent with its known role. Macrophages from mice deficient in macrophage scavenger receptor A (SR-A; CD204) had absent IgM binding and no activation by solid-phase IgM. RNA interference-mediated knockdown of SR-A in human macrophages suppressed activation by solid-phase IgM. IgM binding to SR-A was demonstrated by both co-immunoprecipitation studies and the binding of fluorescently labeled IgM to SR-A-transfected cells. Immunoglobulins on solid-phase particles around macrophages were found in human plaques, increased in ruptured plaques compared with stable ones. These observations indicate that solid-phase IgM and IgG can activate macrophages and destabilize vulnerable plaques. Solid-phase IgM activates macrophages via a novel SR-A pathway.


Subject(s)
Immunoglobulin G/immunology , Immunoglobulin M/immunology , Macrophage Activation/immunology , Plaque, Atherosclerotic/immunology , Scavenger Receptors, Class A/immunology , Animals , Blood Coagulation/physiology , Cells, Cultured , Complement System Proteins/immunology , Coronary Artery Disease/immunology , Cytotoxicity, Immunologic , GPI-Linked Proteins/immunology , Humans , Hydrogen Peroxide/metabolism , Lipoproteins, LDL/metabolism , Mice , Muscle, Smooth, Vascular/immunology , NF-kappa B/physiology , Protein Denaturation , Receptors, Fc/immunology , Receptors, IgG/immunology , Signal Transduction/immunology , Thromboplastin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL