Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
2.
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
3.
Clin Exp Rheumatol ; 32(3 Suppl 82): S11-8, 2014.
Article in English | MEDLINE | ID: mdl-24093733

ABSTRACT

OBJECTIVES: We analysed a large cohort of patients with Takayasu arteritis, seeking robust clinical evidence for prolonged responses to tumour necrosis factor-α (TNF-α) and interleukin-6 receptor (IL-6R) antagonists in severe refractory disease. METHODS: Case notes from ninety-eight patients with Takayasu arteritis were retrospectively reviewed. Drug treatment, laboratory and serial non-invasive imaging data were analysed, and the Indian Takayasu arteritis activity (ITAS) and damage scores (TADs) calculated. RESULTS: Nine patients were treated with biologic therapies. All had previously received high dose prednisolone and ≥1 conventional immunosuppressant. Five patients had failed cyclophosphamide. The patients prescribed biologics had more extensive arterial injury than the remainder of the cohort and persistent active disease (ITAS range 2-9, CRP 12-206 mg/L, TADs 3--1). Eight patients were prescribed anti-TNF-α therapy, three IL-6R blockade. The mean duration of anti-TNF-α treatment was 42 months (maximum 8 years). One patient developed new arterial stenoses while receiving anti-TNF-α and subsequently achieved disease remission with tocilizumab. Two patients have now demonstrated sustained responses to IL-6R inhibition at 19 and 20 months. Following introduction of biologic therapy, serial non-invasive imaging has revealed no significant progression in arterial injury. A significant fall in CRP (p<0.01), prednisolone dose (p<0.01) and ITAS (p<0.01) was observed, with no increase in TADs. CONCLUSIONS: We report for the first time sustained responses to both anti-TNF-α and IL6R antagonists in refractory Takayasu arteritis. As 5/9 patients were cyclophosphamide non-responders, we propose that biologics should now be considered ahead of cyclophosphamide in these young patients.


Subject(s)
Antibodies, Monoclonal, Humanized , Arterial Occlusive Diseases/prevention & control , Receptors, Interleukin-6/antagonists & inhibitors , Takayasu Arteritis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adolescent , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/immunology , Biological Therapy/methods , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Disease Progression , Drug Monitoring , Drug Resistance , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Magnetic Resonance Angiography/methods , Male , Prednisone/administration & dosage , Prednisone/adverse effects , Retrospective Studies , Severity of Illness Index , Takayasu Arteritis/complications , Takayasu Arteritis/drug therapy , Takayasu Arteritis/epidemiology , Takayasu Arteritis/immunology , Takayasu Arteritis/physiopathology , Time Factors , Treatment Outcome , United Kingdom/epidemiology
4.
Biochem J ; 447(2): 193-204, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22849349

ABSTRACT

Vascular endothelial injury predisposes to endothelial dysfunction and atherogenesis. We have investigated the hypothesis that PKCε (protein kinase Cε) is an important upstream regulator of cytoprotective pathways in vascular ECs (endothelial cells). Depletion of PKCε in human ECs reduced expression of the cytoprotective genes A1, A20 and Bcl-2. Conversely, constitutively active PKCε expressed in human ECs increased mRNA and protein levels of these cytoprotective genes, with up-regulation dependent upon ERK1/2 (extracellular-signal-regulated kinase 1/2) activation. Furthermore, inhibition of NF-κB (nuclear factor κB) by the pharmacological antagonist BAY 11-7085 or an IκB (inhibitor of NF-κB) SuperRepressor prevented cytoprotective gene induction. Activation of PKCε enhanced p65 NF-κB DNA binding and elevated NF-κB transcriptional activity. Importantly, although NF-κB activation by PKCε induced cytoprotective genes, it did not up-regulate pro-inflammatory NF-κB targets [E-selectin, VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1)]. Indeed, PKCε exhibited cytoprotective and anti-inflammatory actions, including inhibition of TNFα (tumour necrosis factor α)-induced JNK (c-Jun N-terminal kinase) phosphorylation and ICAM-1 up-regulation, a response attenuated by depletion of A20. Thus we conclude that PKCε plays an essential role in endothelial homoeostasis, acting as an upstream co-ordinator of gene expression through activation of ERK1/2, inhibition of JNK and diversion of the NF-κB pathway to cytoprotective gene induction, and propose that PKCε represents a novel therapeutic target for endothelial dysfunction.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Protein Kinase C-epsilon/metabolism , Animals , DNA-Binding Proteins/biosynthesis , Enzyme Activation , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , Mice , Minor Histocompatibility Antigens , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Tumor Necrosis Factor alpha-Induced Protein 3 , Vascular Cell Adhesion Molecule-1/biosynthesis
5.
Blood ; 113(7): 1598-607, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19036700

ABSTRACT

Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). These end-products are responsible for much of the biologic activity of HO-1, including anti-inflammatory, antiapo-ptotic, antiproliferative, and antioxidant effects. We have identified an additional cytoprotective action, the regulation of complement activation, mediated via induction of decay-accelerating factor (DAF). Pharmacologic inhibition or short-interfering RNA (siRNA) depletion of HO-1 prevented induction of DAF expression in human endothelial cells. In contrast, HO-1 agonists hemin and cobalt protoporphyrin IX significantly increased DAF protein expression, reflecting an increase in transcription and steady-state mRNA. Adenoviral-mediated overexpression of HO-1 increased DAF expression, enhancing protection against C3 deposition and complement-mediated lysis, and this was reversed by DAF inhibitory monoclonal antibody (mAb) 1H4. Likewise, bilirubin, Fe chelation, and overexpression of heavy-chain ferritin all induced DAF expression in endothelial cells (EC). Analysis of cardiac endothelial cells isolated from Hmox1(-/-) mice revealed a 60% reduction in DAF expression compared with Hmox1(+/+) EC, and Hmox1(-/-) cells showed enhanced sensitivity to complement. We propose that modulation of complement activation through induction of DAF represents an important component of the cytoprotective effects of HO-1 against vascular injury, such as that associated with posttransplant vasculopathy, allograft rejection, and ischemia reperfusion.


Subject(s)
Bilirubin/metabolism , CD55 Antigens/metabolism , Complement System Proteins/immunology , Endothelial Cells/immunology , Ferritins/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Animals , CD55 Antigens/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation/physiology , Heme Oxygenase-1/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Protoporphyrins/pharmacology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Umbilical Veins/cytology , Vascular Diseases/immunology , Vascular Diseases/metabolism
6.
Arterioscler Thromb Vasc Biol ; 29(11): 1851-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19729611

ABSTRACT

OBJECTIVE: Proinflammatory mediators influence atherosclerosis by inducing adhesion molecules (eg, VCAM-1) on endothelial cells (ECs) via signaling intermediaries including p38 MAP kinase. Regions of arteries exposed to high shear stress are protected from inflammation and atherosclerosis, whereas low-shear regions are susceptible. Here we investigated whether the transcription factor Nrf2 regulates EC activation in arteries. METHODS AND RESULTS: En face staining revealed that Nrf2 was activated in ECs at an atheroprotected region of the murine aorta where it negatively regulated p38-VCAM-1 signaling, but was expressed in an inactive form in ECs at an atherosusceptible site. Treatment with sulforaphane, a dietary antioxidant, activated Nrf2 and suppressed p38-VCAM-1 signaling at the susceptible site in wild-type but not Nrf2(-/-) animals, indicating that it suppresses EC activation via Nrf2. Studies of cultured ECs revealed that Nrf2 inactivates p38 by suppressing an upstream activator MKK3/6 and by enhancing the activity of the negative regulator MKP-1. CONCLUSIONS: Nrf2 prevents ECs at the atheroprotected site from exhibiting a proinflammatory state via the suppression of p38-VCAM-1 signaling. Pharmacological activation of Nrf2 reduces EC activation at atherosusceptible sites and may provide a novel therapeutic strategy to prevent or reduce atherosclerosis.


Subject(s)
Arteries/enzymology , Arteritis/prevention & control , NF-E2-Related Factor 2/metabolism , Thiocyanates/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Arteries/physiopathology , Arteritis/metabolism , Cells, Cultured/cytology , Cells, Cultured/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation , Inflammation Mediators/metabolism , Isothiocyanates , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/drug effects , Phosphorylation/physiology , Random Allocation , Sensitivity and Specificity , Shear Strength , Signal Transduction , Sulfoxides , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/genetics
8.
Clin Med (Lond) ; 18(2): 180-182, 2018 03.
Article in English | MEDLINE | ID: mdl-29626027

ABSTRACT

A 73-year-old man presented with bilateral leg pain and swelling, and no history of trauma or bleeding disorders. Clinical examination, biochemistry and magnetic resonance imaging of the thighs were suggestive of muscle haematomas. These progressed significantly during the admission, requiring blood transfusion. Normal vascular anatomy on computed tomography, renal and liver function, and absence of infection made a bleeding diathesis more likely. This may be caused by coagulation defects, platelet disorders and vascular fragility. An undetectable serum ascorbic acid level confirmed the clinical suspicion of scurvy, and administration of vitamin C resulted in rapid improvement. Our case provides a structured approach to the diagnosis of bleeding disorders and scurvy, a treatable and potentially fatal disease which is often forgotten.


Subject(s)
Hematoma , Scurvy , Aged , Ascorbic Acid/blood , Hematoma/etiology , Hematoma/pathology , Hematoma/physiopathology , Humans , Male , Scurvy/complications , Scurvy/diagnosis , Scurvy/pathology , Scurvy/physiopathology
9.
Chest ; 129(3): 800-4, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16537885

ABSTRACT

Glucose-insulin and potassium (GIK) infusions are beneficial in treating ischemic myocardial depression. Myocardial depression is also an important feature in septic shock. We describe two cases of pressor-resistant hypodynamic septic shock that responded to high-dose GIK infusions. In each case, hemodynamic profiles improved sufficiently to allow withdrawal of vasopressor agents. Further assessment of GIK in patients with hypodynamic septic shock is necessary to confirm efficacy and prognostic significance.


Subject(s)
Cardioplegic Solutions/therapeutic use , Shock, Septic/drug therapy , Adult , Cardiac Output/drug effects , Epinephrine/administration & dosage , Fatal Outcome , Female , Glucose/therapeutic use , Hemodynamics/drug effects , Humans , Infusions, Intravenous , Insulin/therapeutic use , Male , Middle Aged , Potassium/therapeutic use , Vasoconstrictor Agents/administration & dosage
10.
BMJ Case Rep ; 20152015 Apr 28.
Article in English | MEDLINE | ID: mdl-25920738

ABSTRACT

An 81-year-old South Asian man normally resident in the UK presented with night sweats for over 2 months on a background of weight loss of 4 kg in 6 months. His medical history was significant for metastatic renal cell carcinoma treated 5 years previously with cytoreductive nephrectomy and adjuvant chemotherapy. Following an abnormal chest radiograph showing an ill-defined right paratracheal mass, a CT scan showed eggshell calcification (circumferential calcification) of enlarged low right paratracheal lymph nodes. An endobronchial ultrasound-guided transbronchial needle aspiration of an involved large paratracheal lymph node showed cytology consistent with metastatic renal cell carcinoma.


Subject(s)
Calcinosis/etiology , Carcinoma, Renal Cell/secondary , Kidney Neoplasms/pathology , Lymph Nodes/pathology , Mediastinum/pathology , Aged, 80 and over , Calcinosis/diagnostic imaging , Carcinoma, Renal Cell/diagnostic imaging , Humans , Kidney Neoplasms/diagnostic imaging , Lymphatic Metastasis/diagnosis , Male , Mediastinum/diagnostic imaging , Sweating , Tomography, X-Ray Computed , Treatment Outcome , Weight Loss
11.
Cardiovasc Res ; 106(3): 509-19, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25883219

ABSTRACT

AIMS: Vascular injury leading to endothelial dysfunction is a characteristic feature of chronic renal disease, diabetes mellitus, and systemic inflammatory conditions, and predisposes to apoptosis and atherogenesis. Thus, endothelial dysfunction represents a potential therapeutic target for atherosclerosis prevention. The observation that activity of either protein kinase C epsilon (PKCε) or haem oxygenase-1 (HO-1) enhances endothelial cell (EC) resistance to inflammation and apoptosis led us to test the hypothesis that HO-1 is a downstream target of PKCε. METHODS AND RESULTS: Expression of constitutively active PKCε in human EC significantly increased HO-1 mRNA and protein, whereas conversely aortas or cardiac EC from PKCε-deficient mice exhibited reduced HO-1 when compared with wild-type littermates. Angiotensin II activated PKCε and induced HO-1 via a PKCε-dependent pathway. PKCε activation significantly attenuated TNFα-induced intercellular adhesion molecule-1, and increased resistance to serum starvation-induced apoptosis. These responses were reversed by the HO antagonist zinc protoporphyrin IX. Phosphokinase antibody array analysis identified CREB1((Ser133)) phosphorylation as a PKCε signalling intermediary, and cAMP response element-binding protein 1 (CREB1) siRNA abrogated PKCε-induced HO-1 up-regulation. Likewise, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) was identified as a PKCε target using nuclear translocation and DNA-binding assays, and Nrf2 siRNA prevented PKCε-mediated HO-1 induction. Moreover, depletion of CREB1 inhibited PKCε-induced Nrf2 DNA binding, suggestive of transcriptional co-operation between CREB1 and Nrf2. CONCLUSIONS: PKCε activity in the vascular endothelium regulates HO-1 via a pathway requiring CREB1 and Nrf2. Given the potent protective actions of HO-1, we propose that this mechanism is an important contributor to the emerging role of PKCε in the maintenance of endothelial homeostasis and resistance to injury.


Subject(s)
Apoptosis , Cyclic AMP Response Element-Binding Protein/metabolism , Endothelial Cells/enzymology , Heme Oxygenase-1/biosynthesis , Inflammation/prevention & control , Membrane Proteins/biosynthesis , NF-E2-Related Factor 2/metabolism , Protein Kinase C-epsilon/metabolism , Signal Transduction , Angiotensin II/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Enzyme Induction , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Phosphorylation , Protein Kinase C-epsilon/genetics , Protoporphyrins/pharmacology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transcription, Genetic , Transfection
12.
Free Radic Biol Med ; 48(8): 1013-23, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20083195

ABSTRACT

Although nonsteroidal anti-inflammatory drugs (NSAIDs) provide important control of pain and inflammation, they have been overshadowed by concerns regarding atherothrombotic complications. However, celecoxib seems to have a relatively good cardiovascular profile and may improve endothelial function in coronary heart disease. This led us to the hypothesis that celecoxib induces the vasculoprotective enzyme heme oxygenase-1 (HO-1). In human umbilical vein and aortic endothelial cells, 24-48 h treatment with celecoxib induced HO-1 mRNA and protein expression and increased HO-1 enzyme activity. This effect was not seen with rofecoxib or indomethacin. Supplementation of culture medium with iloprost or prostaglandin E(2) failed to reverse celecoxib-mediated HO-1 induction, indicating a cyclooxygenase-independent mechanism. Rather, this action of celecoxib involved generation of mitochondria-derived reactive oxygen species, Akt phosphorylation, and nuclear translocation of the transcription factor Nrf2, with N-acetylcysteine, PI-3K antagonist LY290042, and dominant-negative Akt abrogating the effects. Furthermore, celecoxib-induced HO-1 was inhibited by dominant-negative Nrf2. The functional significance of HO-1 induction was revealed by celecoxib-mediated inhibition of VCAM-1 expression, a response reversed by the HO-1 antagonist zinc protoporphyrin. HO-1 induction provides a molecular mechanism for clinical observations indicating relative freedom from atherothrombotic complications in patients taking celecoxib compared to other NSAIDs with comparable anti-inflammatory activity.


Subject(s)
Heme Oxygenase-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Acetylcysteine/pharmacology , Celecoxib , Chromones/pharmacology , Endothelial Cells/drug effects , Enzyme Induction , Humans , Lactones/pharmacology , Morpholines/pharmacology , NF-E2-Related Factor 2/pharmacology , Oxidation-Reduction , Protein Transport/drug effects , Protoporphyrins/pharmacology , Signal Transduction/drug effects , Sulfones/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/biosynthesis
13.
Cardiovasc Res ; 85(4): 701-10, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19903700

ABSTRACT

AIMS: Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily of ligand-activated transcriptional regulators. PPARdelta has an established role in metabolism, wound healing, and angiogenesis. However, little is known about its function in endothelial homeostasis. We investigated the role of PPARdelta and its co-activator, PPARgamma co-activator 1alpha (PGC1alpha), in vasculoprotection against oxidant-induced injury via induction of haem oxygenase-1. METHODS AND RESULTS: En face confocal microscopy of murine aortas demonstrated that the PPARdelta-selective ligand GW501516 induced endothelial haem oxygenase-1 expression. In vitro PPARdelta ligands induced a significant increase in haem oxygenase-1 mRNA, protein, and enzyme activity, resulting in enhanced human endothelial cell protection against cellular stress induced by hydrogen peroxide or leptin. Moreover, adenoviral-mediated overexpression of haem oxygenase-1 increased PPARdelta promoter activity and mRNA levels, amplifying the effect of PPARdelta ligands through a positive feedback loop. Mutation of PPAR response element binding sites in the haem oxygenase-1 promoter/enhancer region revealed haem oxygenase-1 to be a direct PPARdelta target gene. Inhibition of either haem oxygenase-1 or PPARdelta abrogated PPARdelta ligand-induced endothelial cytoprotection. Furthermore, siRNA depletion of PGC1alpha demonstrated that this co-regulator acts as an essential PPARdelta transcriptional co-activator for haem oxygenase-1 induction by PPARdelta ligands and its subsequent cytoprotective actions. CONCLUSION: We have identified an important relationship between PPARdelta, PGC1alpha, and haem oxygenase-1, demonstrating that haem oxygenase-1 induction plays an important role in cytoprotective actions of PPARdelta ligands in vascular endothelium. In light of the protective effects of haem oxygenase-1 against atherogenesis, we suggest that PPARdelta represents a potentially important therapeutic target in the vasculature.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Heat-Shock Proteins/metabolism , Heme Oxygenase-1/metabolism , Oxidative Stress/physiology , PPAR delta/metabolism , Transcription Factors/metabolism , Animals , Aorta/cytology , Apoptosis/physiology , Atherosclerosis/pathology , Cells, Cultured , Endothelial Cells/cytology , Enzyme Activation/physiology , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Heat-Shock Proteins/genetics , Heme Oxygenase-1/genetics , Homeostasis/drug effects , Homeostasis/physiology , Humans , Ligands , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , PPAR delta/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Small Interfering , Thiazoles/pharmacology , Transcription Factors/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Umbilical Veins/cytology
14.
J Biol Chem ; 284(28): 18882-92, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19457866

ABSTRACT

In addition to cholesterol-lowering properties, statins exhibit lipid-independent immunomodulatory, anti-inflammatory actions. However, high concentrations are typically required to induce these effects in vitro, raising questions concerning therapeutic relevance. We present evidence that endothelial cell sensitivity to statins depends upon shear stress. Using heme oxygenase-1 expression as a model, we demonstrate differential heme oxygenase-1 induction by atorvastatin in atheroresistant compared with atheroprone sites of the murine aorta. In vitro, exposure of human endothelial cells to laminar shear stress significantly reduced the statin concentration required to induce heme oxygenase-1 and protect against H(2)O(2)-mediated injury. Synergy was observed between laminar shear stress and atorvastatin, resulting in optimal expression of heme oxygenase-1 and resistance to oxidative stress, a response inhibited by heme oxygenase-1 small interfering RNA. Moreover, treatment of laminar shear stress-exposed endothelial cells resulted in a significant fall in intracellular cholesterol. Mechanistically, synergy required Akt phosphorylation, activation of Kruppel-like factor 2, NF-E2-related factor-2 (Nrf2), increased nitric-oxide synthase activity, and enhanced HO-1 mRNA stability. In contrast, heme oxygenase-1 induction by atorvastatin in endothelial cells exposed to oscillatory flow was markedly attenuated. We have identified a novel relationship between laminar shear stress and statins, demonstrating that atorvastatin-mediated heme oxygenase-1-dependent antioxidant effects are laminar shear stress-dependent, proving the principle that biomechanical signaling contributes significantly to endothelial responsiveness to pharmacological agents. Our findings suggest statin pleiotropy may be suboptimal at disturbed flow atherosusceptible sites, emphasizing the need for more specific therapeutic agents, such as those targeting Kruppel-like factor 2 or Nrf2.


Subject(s)
Endothelium, Vascular/metabolism , Heme Oxygenase-1/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Adenoviridae/metabolism , Animals , Biomechanical Phenomena , Endothelium, Vascular/cytology , Heme Oxygenase-1/metabolism , Humans , Hydrogen Peroxide/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phosphorylation , RNA, Small Interfering/metabolism , Stress, Mechanical
16.
J Biol Chem ; 283(21): 14636-44, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18362151

ABSTRACT

Complement activation may predispose to vascular injury and atherogenesis. The atheroprotective actions of unidirectional laminar shear stress led us to explore its influence on endothelial cell expression of complement inhibitory proteins CD59 and decay-accelerating factor. Human umbilical vein and aortic endothelial cells were exposed to laminar shear stress (12 dynes/cm(2)) or disturbed flow (+/- 5 dynes/cm(2) at 1Hz) in a parallel plate flow chamber. Laminar shear induced a flow rate-dependent increase in steady-state CD59 mRNA, reaching 4-fold at 12 dynes/cm(2). Following 24-48 h of laminar shear stress, cell surface expression of CD59 was up-regulated by 100%, whereas decay-accelerating factor expression was unchanged. The increase in CD59 following laminar shear was functionally significant, reducing C9 deposition and complement-mediated lysis of flow-conditioned endothelial cells by 50%. Although CD59 induction was independent of PI3-K, ERK1/2 and nitric oxide, an RNA interference approach demonstrated dependence upon an ERK5/KLF2 signaling pathway. In contrast to laminar shear stress, disturbed flow failed to induce endothelial cell CD59 protein expression. Likewise, CD59 expression on vascular endothelium was significantly higher in atheroresistant regions of the murine aorta exposed to unidirectional laminar shear stress, when compared with atheroprone areas exposed to disturbed flow. We propose that up-regulation of CD59 via ERK5/KLF2 activation leads to endothelial resistance to complement-mediated injury and protects from atherogenesis in regions of laminar shear stress.


Subject(s)
Aorta/injuries , Aorta/metabolism , CD59 Antigens/metabolism , Cytoprotection , Kruppel-Like Transcription Factors/metabolism , Umbilical Cord/metabolism , Animals , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Signal Transduction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL