Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrea Bauer is active.

Publication


Featured researches published by Andrea Bauer.


Journal of Biological Chemistry | 2009

Induction of the Cytoprotective Enzyme Heme Oxygenase-1 by Statins Is Enhanced in Vascular Endothelium Exposed to Laminar Shear Stress and Impaired by Disturbed Flow

Faisal Ali; Mustafa Zakkar; Kersti Karu; Elaine A. Lidington; Shahir S. Hamdulay; Joseph J. Boyle; Mire Zloh; Andrea Bauer; Dorian O. Haskard; Paul C. Evans; Justin C. Mason

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 H2O2-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.


Cardiovascular Research | 2010

PPARδ and PGC1α act cooperatively to induce haem oxygenase-1 and enhance vascular endothelial cell resistance to stress

Faisal Ali; Nadira Ali; Andrea Bauer; Joseph J. Boyle; Shahir S. Hamdulay; Dorian O. Haskard; Anna M. Randi; Justin C. Mason

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.


Cardiovascular Research | 2015

PKCε-CREB-Nrf2 signalling induces HO-1 in the vascular endothelium and enhances resistance to inflammation and apoptosis.

Hayley Mylroie; Odile Dumont; Andrea Bauer; Clare Thornton; John Mackey; Damien Calay; Shahir S. Hamdulay; Joan R. Choo; Joseph J. Boyle; Allen M. Samarel; Anna M. Randi; Paul C. Evans; Justin C. Mason

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.


Annals of the Rheumatic Diseases | 2016

Methotrexate-mediated activation of an AMPK-CREB-dependent pathway: a novel mechanism for vascular protection in chronic systemic inflammation

Clare Thornton; Fahad Al-Rashed; Damien Calay; Graeme M. Birdsey; Andrea Bauer; Hayley Mylroie; Bernard J Morley; Anna M. Randi; Dorian O. Haskard; Joseph J. Boyle; Justin C. Mason

Aims Premature cardiovascular events complicate chronic inflammatory conditions. Low-dose weekly methotrexate (MTX), the most widely used disease-modifying drug for rheumatoid arthritis (RA), reduces disease-associated cardiovascular mortality. MTX increases intracellular accumulation of adenosine monophosphate (AMP) and 5-aminoimidazole-4-carboxamide ribonucleotide which activates AMP-activated protein kinase (AMPK). We hypothesised that MTX specifically protects the vascular endothelium against inflammatory injury via induction of AMPK-regulated protective genes. Methods/results In the (NZW×BXSB)F1 murine model of inflammatory vasculopathy, MTX 1 mg/kg/week significantly reduced intramyocardial vasculopathy and attenuated end-organ damage. Studies of human umbilical vein endothelial cells (HUVEC) and arterial endothelial cells (HAEC) showed that therapeutically relevant concentrations of MTX phosphorylate AMPKαThr172, and induce cytoprotective genes including manganese superoxide dismutase (MnSOD) and haem oxygenase-1 (HO-1). These responses were preserved when HUVECs were pretreated with tumour necrosis factor-α to mimic dysfunctional endothelium. Furthermore, MTX protected against glucose deprivation-induced endothelial apoptosis. Mechanistically, MTX treatment led to cyclic AMP response element-binding protein (CREB)Ser133 phosphorylation, while AMPK depletion attenuated this response and the induction of MnSOD and HO-1. CREB siRNA inhibited upregulation of both cytoprotective genes by MTX, while chromatin immunoprecipitation demonstrated CREB binding to the MnSOD promoter in MTX-treated EC. Likewise, treatment of (NZW×BXSB)F1 mice with MTX enhanced AMPKαThr172 phosphorylation and MnSOD, and reduced aortic intercellular adhesion molecule-1 expression. Conclusions These data suggest that MTX therapeutically conditions vascular endothelium via activation of AMPK-CREB. We propose that this mechanism contributes to the protection against cardiovascular events seen in patients with RA treated with MTX.


Biochemical Journal | 2012

Protein kinase Cε activity induces anti-inflammatory and anti-apoptotic genes via an ERK1/2- and NF-κB-dependent pathway to enhance vascular protection.

Odile Dumont; Hayley Mylroie; Andrea Bauer; Damien Calay; Andrea Sperone; Clare Thornton; Shahir S. Hamdulay; Nadira Ali; Joseph J. Boyle; Joan R. Choo; Allen M. Samarel; Dorian O. Haskard; Anna M. Randi; Paul C. Evans; Justin C. Mason

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.


Scientific Reports | 2016

Identification of cyclins A1, E1 and vimentin as downstream targets of heme oxygenase-1 in vascular endothelial growth factor-mediated angiogenesis

Andrea Bauer; Hayley Mylroie; Clare Thornton; Damien Calay; Graeme M. Birdsey; Allan P. Kiprianos; Garrick K. Wilson; Miguel P. Soares; Xiaoke Yin; Manuel Mayr; Anna M. Randi; Justin C. Mason

Angiogenesis is an essential physiological process and an important factor in disease pathogenesis. However, its exploitation as a clinical target has achieved limited success and novel molecular targets are required. Although heme oxygenase-1 (HO-1) acts downstream of vascular endothelial growth factor (VEGF) to modulate angiogenesis, knowledge of the mechanisms involved remains limited. We set out identify novel HO-1 targets involved in angiogenesis. HO-1 depletion attenuated VEGF-induced human endothelial cell (EC) proliferation and tube formation. The latter response suggested a role for HO-1 in EC migration, and indeed HO-1 siRNA negatively affected directional migration of EC towards VEGF; a phenotype reversed by HO-1 over-expression. EC from Hmox1−/− mice behaved similarly. Microarray analysis of HO-1-depleted and control EC exposed to VEGF identified cyclins A1 and E1 as HO-1 targets. Migrating HO-1-deficient EC showed increased p27, reduced cyclin A1 and attenuated cyclin-dependent kinase 2 activity. In vivo, cyclin A1 siRNA inhibited VEGF-driven angiogenesis, a response reversed by Ad-HO-1. Proteomics identified structural protein vimentin as an additional VEGF-HO-1 target. HO-1 depletion inhibited VEGF-induced calpain activity and vimentin cleavage, while vimentin silencing attenuated HO-1-driven proliferation. Thus, vimentin and cyclins A1 and E1 represent VEGF-activated HO-1-dependent targets important for VEGF-driven angiogenesis.


Scientific Reports | 2018

Celecoxib exerts protective effects in the vascular endothelium via COX-2-independent activation of AMPK-CREB-Nrf2 signalling

Fahad Al-Rashed; Damien Calay; Marie Lang; Clare Thornton; Andrea Bauer; Allan P. Kiprianos; Dorian O. Haskard; Anusha Seneviratne; Joseph J. Boyle; Alex H. Schönthal; Caroline P.D. Wheeler-Jones; Justin C. Mason

Although concern remains about the athero-thrombotic risk posed by cyclo-oxygenase (COX)-2-selective inhibitors, recent data implicates rofecoxib, while celecoxib appears equivalent to NSAIDs naproxen and ibuprofen. We investigated the hypothesis that celecoxib activates AMP kinase (AMPK) signalling to enhance vascular endothelial protection. In human arterial and venous endothelial cells (EC), and in contrast to ibuprofen and naproxen, celecoxib induced the protective protein heme oxygenase-1 (HO-1). Celecoxib derivative 2,5-dimethyl-celecoxib (DMC) which lacks COX-2 inhibition also upregulated HO-1, implicating a COX-2-independent mechanism. Celecoxib activated AMPKα(Thr172) and CREB-1(Ser133) phosphorylation leading to Nrf2 nuclear translocation. Importantly, these responses were not reproduced by ibuprofen or naproxen, while AMPKα silencing abrogated celecoxib-mediated CREB and Nrf2 activation. Moreover, celecoxib induced H-ferritin via the same pathway, and increased HO-1 and H-ferritin in the aortic endothelium of mice fed celecoxib (1000 ppm) or control chow. Functionally, celecoxib inhibited TNF-α-induced NF-κB p65(Ser536) phosphorylation by activating AMPK. This attenuated VCAM-1 upregulation via induction of HO-1, a response reproduced by DMC but not ibuprofen or naproxen. Similarly, celecoxib prevented IL-1β-mediated induction of IL-6. Celecoxib enhances vascular protection via AMPK-CREB-Nrf2 signalling, a mechanism which may mitigate cardiovascular risk in patients prescribed celecoxib. Understanding NSAID heterogeneity and COX-2-independent signalling will ultimately lead to safer anti-inflammatory drugs.


Annals of the Rheumatic Diseases | 2017

THU0053 Selective activation of an AMPK-CREB-NRF2-dependent pathway by celecoxib induces vasculoprotective genes and mitigates against cardiovascular risk

Fahad Al-Rashed; Damien Calay; Clare Thornton; Andrea Bauer; A Kiprianos; Dorian O. Haskard; A Seneviratne; Joseph J. Boyle; Justin C. Mason

Background Although concern remains about the athero-thrombotic risk posed by COX-2-selective inhibitors (COXIBs), the recent PRECISION trial demonstrated non-inferiority of moderate dose celecoxib when compared to naproxen and ibuprofen with respect to cardiovascular safety, with fewer actual CV events recorded for celecoxib. Moreover, celecoxib proved significantly safer than either comparator in regard to gastrointestinal events1. Given the markedly different cardiovascular risk associated with celecoxib and rofecoxib, we investigated the hypothesis that, in addition to cyclo-oxygenase inhibition, celecoxib specifically activates COX-2-independent AMP kinase (AMPK) signalling to exert protective effects in the vascular endothelium. Objectives To investigate COX-2-independent vasculoprotective signalling pathways activated by celecoxib in human endothelium. Methods In vitro studies of celecoxib, rofecoxib, ibuprofen and naproxen were performed on human umbilical vein and human aortic endothelial cells (HUVEC and HAEC). Inhibition of signalling pathways was achieved using siRNA. The vascular effects of celecoxib in vivo were studied in C57Bl/6 mice fed celecoxib (1000 ppm) or control chow (48 hrs). Aortic tissue was snap-frozen and sections studied by immunofluorescence confocal microscopy. Results At therapeutically relevant concentrations celecoxib (1–10μ M) induced the vasculoprotective protein heme oxygenase-1 (HO-1) in HUVEC and HAEC (EC) (p<0.01). In contrast, rofecoxib and the commonly used non-selective NSAIDs ibuprofen and naproxen failed to induce HO-1. Celecoxib derivative 2,5-dimethyl-celecoxib (DMC), which lacks COX-2 inhibition, also upregulated HO-1, implicating a COX-2-independent mechanism. Immunoblotting demonstrated that celecoxib and DMC induce AMPKα(Thr172) and CREB-1(Ser133) phosphorylation leading to Nrf2 nuclear translocation (p<0.05). These responses were not seen with ibuprofen or naproxen, while siRNA depletion of AMPKα abrogated celecoxib-mediated CREB and Nrf2 activation (p<0.05). Acting via the same pathway, celecoxib induced additional cytoprotective genes including H-ferritin. In vivo, celecoxib similarly increased HO-1 and H-ferritin in murine aortic endothelium when compared to control-fed mice (p<0.05). Functionally, celecoxib treatment inhibited TNF-α-induced NF-κB p65(Ser536) phosphorylation by increasing AMPK activity. This attenuated VCAM-1 upregulation via induction of HO-1, as revealed by HO-1 siRNA (p<0.05). Similarly, celecoxib prevented the IL-1-mediated increase of IL-6 mRNA (p<0.01). These responses were not seen with ibuprofen or naproxen. Conclusions Celecoxib induces anti-inflammatory, anti-oxidant proteins HO-1 and H-ferritin in human vascular endothelium via a novel AMPK-CREB-Nrf2-dependent pathway. This mechanism may contribute to the important and marked differences in cardiovascular risk between celecoxib and rofecoxib. Understanding mechanisms underlying NSAID heterogeneity may ultimately lead to the development of safer anti-inflammatory drugs. References Nissen SE et al. Cardiovascular Safety of Celecoxib, Naproxen, or Ibuprofen for Arthritis. N Engl J Med. 2016;375:2519–29. Disclosure of Interest None declared


Biochemistry | 1998

STRUCTURE-ACTIVITY RELATIONSHIP OF SYNTHETIC PHOSPHOINOSITOLGLYCANS MIMICKING METABOLIC INSULIN ACTION

Wendelin Frick; Andrea Bauer; Jochen Bauer; Susanne Wied; Günter Müller


Biochemical Journal | 1998

Insulin-mimetic signalling of synthetic phosphoinositolglycans in isolated rat adipocytes

Wendelin Frick; Andrea Bauer; Jochen Bauer; Susanne Wied; Günter Müller

Collaboration


Dive into the Andrea Bauer's collaboration.

Top Co-Authors

Avatar

Justin C. Mason

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dorian O. Haskard

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna M. Randi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Clare Thornton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Damien Calay

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hayley Mylroie

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shahir S. Hamdulay

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Fahad Al-Rashed

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Graeme M. Birdsey

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge