Network


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

Hotspot


Dive into the research topics where Ersilia Cipolletta is active.

Publication


Featured researches published by Ersilia Cipolletta.


Journal of the American Heart Association | 2012

CaMK4 Gene Deletion Induces Hypertension

Gaetano Santulli; Ersilia Cipolletta; Daniela Sorriento; Carmine Del Giudice; Antonio Anastasio; Sara Monaco; Angela Serena Maione; Gianluigi Condorelli; Annibale Alessandro Puca; Bruno Trimarco; Maddalena Illario; Guido Iaccarino

Background The expression of calcium/calmodulin-dependent kinase IV (CaMKIV) was hitherto thought to be confined to the nervous system. However, a recent genome-wide analysis indicated an association between hypertension and a single-nucleotide polymorphism (rs10491334) of the human CaMKIV gene (CaMK4), which suggests a role for this kinase in the regulation of vascular tone. Methods and Results To directly assess the role of CaMKIV in hypertension, we characterized the cardiovascular phenotype of CaMK4−/− mice. They displayed a typical hypertensive phenotype, including high blood pressure levels, cardiac hypertrophy, vascular and kidney damage, and reduced tolerance to chronic ischemia and myocardial infarction compared with wild-type littermates. Interestingly, in vitro experiments showed the ability of this kinase to activate endothelial nitric oxide synthase. Eventually, in a population study, we found that the rs10491334 variant associates with a reduction in the expression levels of CaMKIV in lymphocytes from hypertensive patients. Conclusions Taken together, our results provide evidence that CaMKIV plays a pivotal role in blood pressure regulation through the control of endothelial nitric oxide synthase activity. (J Am Heart Assoc. 2012;1:e001081 doi: 10.1161/JAHA.112.001081.)


Circulation Research | 2005

Ischemic Neoangiogenesis Enhanced by β2-Adrenergic Receptor Overexpression: A Novel Role for the Endothelial Adrenergic System

Guido Iaccarino; Michele Ciccarelli; Daniela Sorriento; Gennaro Galasso; Alfonso Campanile; Gaetano Santulli; Ersilia Cipolletta; Vincenzo Cerullo; Vincenzo Cimini; Giovanna Giuseppina Altobelli; Federico Piscione; Ornella Priante; Lucio Pastore; Massimo Chiariello; F. Salvatore; Walter J. Koch; Bruno Trimarco

&bgr;2-Adrenergic receptors (&bgr;2ARs) are widely expressed, although their physiological relevance in many tissues is not yet fully understood. In vascular endothelial cells, they regulate NO release and vessel tone. Here we provide novel evidence that &bgr;2ARs can regulate neoangiogenesis in response to chronic ischemia. We used in vivo adenoviral-mediated gene transfer of the human &bgr;2AR to the endothelium of the rat femoral artery and increased &bgr;2AR signaling resulting in ameliorated angiographic blood flow and hindlimb perfusion after chronic ischemia. Histological analysis confirmed that &bgr;2AR overexpression also produced benefits on capillary density. The same maneuver partially rescued impaired angiogenesis in spontaneously hypertensive rats (SHR), whereas gene delivery of the G-protein–coupling defective mutant Ile164 &bgr;2AR failed to provide ameliorations. Stimulation of endogenous and overexpressed &bgr;2AR on endothelial cells in vitro was found to regulate cell number by inducing proliferation and [3H]-thymidine incorporation through means of extracellular receptor-activated kinase and vascular endothelial growth factor. The &bgr;2AR also has novel effects on endothelial cell number through stimulation of proapoptosis and antiapoptosis pathways involving p38 mitogen-activated protein kinase and PI3-kinase/Akt activation. Therefore, &bgr;2ARs play a critical role in endothelial cell proliferation and function including revascularization, suggesting a novel and physiologically relevant role in neoangiogenesis in response to ischemia.


Cardiovascular Research | 2009

The G protein coupled receptor kinase 2 plays an essential role in beta-adrenergic receptor-induced insulin resistance

Ersilia Cipolletta; Alfonso Campanile; Gaetano Santulli; Emma Sanzari; Dario Leosco; Pietro Campiglia; Bruno Trimarco; Guido Iaccarino

AIMS Insulin (Ins) resistance (IRES) associates to increased cardiovascular risk as observed in metabolic syndrome. Chronic stimulation of beta-adrenergic receptors (betaAR) due to exaggerated sympathetic nervous system activity is involved in the pathogenesis of IRES. The cellular levels of G protein coupled receptor kinase 2 (GRK2) increase during chronic betaAR stimulation, leading to betaAR desensitization. We tested the hypothesis that GRK2 plays a role in betaAR-induced IRES. METHODS AND RESULTS We evaluated Ins-induced glucose uptake and signalling responses in vitro in cell overexpressing the beta(2)AR, the GRK2, or the catalytically dead mutant GRK2-DN. In a model of increased adrenergic activity, IRES and elevated cellular GRK2 levels, the spontaneously hypertensive rats (SHR) we performed the intravenous glucose tolerance test load. To inhibit GRK2, we synthesized a peptide based on the catalytical sequence of GRK2 conjugated with the antennapedia internalization sequence (Ant-124). Ins in human kidney embryonic (HEK-293) cells causes rapid accumulation of GRK2, tyrosine phosphorylation of Ins receptor substrate 1 (IRS1) and induces glucose uptake. In the same cell type, transgenic beta(2)AR overexpression causes GRK2 accumulation associated with significant deficit of IRS1 activation and glucose uptake by Ins. Similarly, transgenic GRK2 overexpression prevents Ins-induced tyrosine phosphorylation of IRS1 and glucose uptake, whereas GRK2-DN ameliorates glucose extraction. By immunoprecipitation, GRK2 binds IRS1 but not the Ins receptor in an Ins-dependent fashion, which is lost in HEK-GRK2 cells. Ant-124 improves Ins-induced glucose uptake in HEK-293 and HEK-GRK2 cells, but does not prevent GRK2/IRS1 interaction. In SHR, Ant-124 infusion for 30 days ameliorates IRES and IRS1 tyrosine phosphorylation. CONCLUSION Our results suggest that GRK2 mediates adrenergic IRES and that inhibition of GRK2 activity leads to increased Ins sensitivity both in cells and in animal model of IRES.


Circulation | 2004

AKT Participates in Endothelial Dysfunction in Hypertension

Guido Iaccarino; Michele Ciccarelli; Daniela Sorriento; Ersilia Cipolletta; Vincenzo Cerullo; Gianni Luigi Iovino; Alessandro Paudice; Andrea Elia; Gaetano Santulli; Alfonso Campanile; Oreste Arcucci; Lucio Pastore; F. Salvatore; Gianluigi Condorelli; Bruno Trimarco

Background—In hypertension, reduced nitric oxide production and blunted endothelial vasorelaxation are observed. It was recently reported that AKT phosphorylates and activates endothelial nitric oxide synthase and that impaired kinase activity may be involved in endothelial dysfunction. Methods and Results—To identify the physiological role of the kinase in normotensive Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR), we used adenoviral vectors to transfer the human AKT1 gene selectively to the common carotid endothelium. In vitro, endothelial vasorelaxations to acetylcholine, isoproterenol, and insulin were blunted in control carotids from SHR compared with WKY rats, and human AKT1 overexpression corrected these responses. Similarly, blood flow assessed in vivo by Doppler ultrasound was reduced in SHR compared with WKY carotids and normalized after AKT1 gene transfer. In primary cultured endothelial cells, we evaluated AKT phosphorylation, activity, and compartmentalization and observed a mislocalization of the kinase in SHR. Conclusions—We conclude that AKT participates in the settings of endothelial dysfunction in SHR rats by impaired membrane localization. Our data suggest that AKT is involved in endothelium dysfunction in hypertension.


Circulation | 2002

β2-Adrenergic Receptor Gene Delivery to the Endothelium Corrects Impaired Adrenergic Vasorelaxation in Hypertension

Guido Iaccarino; Ersilia Cipolletta; Antonia Fiorillo; Mario Annecchiarico; Michele Ciccarelli; Vincenzo Cimini; Walter J. Koch; Bruno Trimarco

Background—Impaired &bgr;-adrenergic receptor (AR)–mediated vasorelaxation in hypertension plays a role in increased peripheral vascular resistance and blood pressure. Because the &bgr;2AR is the most abundant vascular AR subtype, we sought to enhance &bgr;AR vasorelaxation by overexpressing &bgr;2ARs via adenoviral-mediated gene transfer (AD&bgr;2AR) to the vascular endothelium of the carotid artery. Methods and Results—In normotensive Wistar-Kyoto (WKY) and spontaneously hypertensive (SHR) rats, we exposed the right common carotid artery to AD&bgr;2AR in situ for 15 minutes by injection into the lumen while the blood flow was interrupted. Control carotids received an empty vector (ADempty). Three days later, transgene expression and selective endothelial localization were confirmed in infected vessels. Vasoregulation after &bgr;2AR overexpression (2-fold) was studied in isolated organ baths. AD&bgr;2AR carotid responses to &agr;1AR and &agr;2AR agonists were not affected, whereas responses to epinephrine were altered and &bgr;AR-mediated vasorelaxation was enhanced after &bgr;2AR overexpression. As expected, &bgr;AR-mediated vasodilatation in control carotids of SHR rats was significantly less than in similar control WKY carotid arteries. AD&bgr;2AR treatment enhanced &bgr;AR vasorelaxation in SHR to levels similar to those seen in AD&bgr;2AR WKY carotids. Conclusions—Our results demonstrate a critical role for the endothelium in &bgr;AR-mediated vasorelaxation and suggest that impaired &bgr;AR signaling may account for dysfunctional &bgr;AR vasorelaxation in hypertension rather than impaired endothelium-dependent nitric oxide metabolism.


Cellular Signalling | 2012

Mitochondrial localization unveils a novel role for GRK2 in organelle biogenesis

A Fusco; Gaetano Santulli; Daniela Sorriento; Ersilia Cipolletta; Corrado Garbi; Gerald W. Dorn; Bruno Trimarco; Antonio Feliciello; Guido Iaccarino

Metabolic stimuli such as insulin and insulin like growth factor cause cellular accumulation of G protein coupled receptor kinase 2 (GRK2), which in turn is able to induce insulin resistance. Here we show that in fibroblasts, GRK2 is able to increase ATP cellular content by enhancing mitochondrial biogenesis; also, it antagonizes ATP loss after hypoxia/reperfusion. Interestingly, GRK2 is able to localize in the mitochondrial outer membrane, possibly through one region within the RGS homology domain and one region within the catalytic domain. In vivo, GRK2 removal from the skeletal muscle results in reduced ATP production and impaired tolerance to ischemia. Our data show a novel sub-cellular localization of GRK2 in the mitochondria and an unexpected role in regulating mitochondrial biogenesis and ATP generation.


Endocrinology | 2010

Calmodulin-Dependent Kinase II Mediates Vascular Smooth Muscle Cell Proliferation and Is Potentiated by Extracellular Signal Regulated Kinase

Ersilia Cipolletta; Sara Monaco; Angela Serena Maione; L. Vitiello; Pietro Campiglia; Lucio Pastore; Carlo Franchini; E. Novellino; V. Limongelli; K. U. Bayer; Anthony R. Means; Guido Rossi; Bruno Trimarco; Guido Iaccarino; M. Illario

Vascular smooth muscle cell (VSMC) proliferation contributes to vascular remodeling in atherosclerosis and hypertension. Calcium-dependent signaling through calcium/calmodulin-dependent kinase II (CaMKII) and ERK1/2 activation plays an important role in the regulation of VSMC proliferation by agents such as alpha-adrenergic receptor agonists. Nevertheless, how the CaMKII and ERK pathways interact in VSMCs has yet to be characterized. The aim of the present study was to clarify this interaction in response to alpha(1)-adrenergic receptor-mediated VSMC proliferation. We discovered that phenylephrine stimulation resulted in complex formation between CaMKII and ERK in a manner that facilitated phosphorylation of both protein kinases. To assess the effects of CaMKII/ERK association on VSMC proliferation, we inhibited endogenous CaMKII either pharmacologically or by adenoviral-mediated gene transfer of a kinase-inactive CaMKII mutant. Inhibition of CaMKII activation but not CaMKII autonomous activity significantly decreased formation of the CaMKII/ERK complex. On the contrary, the expression of constitutively active CaMKII enhanced VSMC growth and CaMKII/ERK association. In addressing the mechanism of this effect, we found that CaMKII could not directly phosphorylate ERK but instead enhanced Raf1 activation. By contrast, ERK interaction with CaMKII facilitated CaMKII phosphorylation and promoted its nuclear localization. Our results reveal a critical role for CaMKII in VSMC proliferation and imply that CaMKII facilitates assembly of the Raf/MEK/ERK complex and that ERK enhances CaMKII activation and influences its subcellular localization.


Clinical and Translational Science | 2008

Enhanced GRK2 Expression and Desensitization of βAR Vasodilatation in Hypertensive Patients

Raffaele Izzo; Ersilia Cipolletta; Michele Ciccarelli; Alfonso Campanile; Gaetano Santulli; Gianluigi Palumbo; Antonio Vasta; Salvatore Formisano; Bruno Trimarco; Guido Iaccarino

Increased levels of G protein coupled receptor kinase GRK2 appear to participate in hypertension presumably through the desensitization of β adrenergic receptors (βARs) that mediate vasodilatation. There are contrasting data on the occurrence of βAR desensitization in the vasculature, we therefore investigated βAR vasodilatation and desensitization in normotensives and in hypertensive humans. In blood lymphocytes, we assessed βAR signaling and GRK2 expression and found βAR signaling alterations and, consistent with desensitization, ncreased GRK2 levels in hypertensives. We studied in vivo vasodilatation to the βAR agonist isoproterenol (ISO) injected in the brachia artery in control conditions and during the concomitant infusion of heparin, a known in vitro nonspecific GRK inhibitor. ISO induced a dose‐dependent vasorelaxation that was attenuated in hypertensives indicating a loss of βAR signaling. Intra‐arterial infusion of heparin nhibited lymphocyte GRK2 activity and prevented desensitization of βAR vasodilatation in normotensives. In hypertensives, heparin restored vasodilatation to ISO, to levels observed in normotensives. Our results suggest that βAR desensitization does indeed occur at the vascular levels in vivo, and that heparin by acting as a GRK inhibitor prevents this in normotensives and restores impaired βAR vasodilation in hypertensives. We conclude that desensitization participates to impaired βAR vasodilation in hypertension.


Cellular Signalling | 2009

Calcium-calmodulin-dependent kinase II (CaMKII) mediates insulin-stimulated proliferation and glucose uptake.

Maddalena Illario; Sara Monaco; Anna Lina Cavallo; Iolanda Esposito; Pietro Formisano; Luca D'Andrea; Ersilia Cipolletta; Bruno Trimarco; Gianfranco Fenzi; Guido Rossi; Mario Vitale

Cellular growth and glucose uptake are regulated by multiple signals generated by the insulin receptor. The mechanisms of individual modulation of these signals remain somewhat elusive. We investigated the role of CaMKII in insulin signalling in a rat skeletal muscle cell line, demonstrating that CaMKII modulates the insulin action on DNA synthesis and the negative feedback that down regulates glucose uptake. Insulin stimulation generated partly independent signals leading to the rapid activation of Akt, Erk-1/2 and CaMKII. Akt activation was followed by Glut-4 translocation to the plasma membrane and increase of glucose uptake. Then, IRS-1 was phosphorylated at S612, the IRS-1/p85PI3K complex was disrupted, Akt was no more phosphorylated and both Glut-4 translocation and glucose uptake were reduced. Inhibition of CaMKII abrogated the insulin-induced Erk-1/2 activation, DNA synthesis and phosphorylation of IRS-1 at S612. Inhibition of CaMKII also abrogated the down-regulation of insulin-stimulated Akt phosphorylation, Glut-4 membrane translocation and glucose uptake. These results demonstrate that: 1 - CaMKII modulates the insulin-induced Erk-1/2 activation and cell proliferation; 2 - after the initial stimulation of the IRS-1/Akt pathway, CaMKII mediates the down-regulation of stimulated glucose uptake. This represents a novel mechanism in the selective control of insulin signals, and a possible site for pharmacological intervention.


British Journal of Pharmacology | 2011

Impaired neoangiogenesis in β2-adrenoceptor gene-deficient mice: restoration by intravascular human β2-adrenoceptor gene transfer and role of NFκB and CREB transcription factors

Michele Ciccarelli; Daniela Sorriento; Ersilia Cipolletta; Gaetano Santulli; A Fusco; Rui-Hai Zhou; Andrea D. Eckhart; Karsten Peppel; Walter J. Koch; Bruno Trimarco; Guido Iaccarino

There is much evidence supporting the role of β2‐adrenoceptors (β2AR) in angiogenesis but the mechanisms underlying their effects have not been elucidated. Hence, we studied post‐ischaemic angiogenesis in the hindlimb (HL) of β2AR knock‐out mice (β2AR−/−) in vivo and explored possible molecular mechanisms in vitro.

Collaboration


Dive into the Ersilia Cipolletta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruno Trimarco

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Daniela Sorriento

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Gaetano Santulli

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raffaele Izzo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Alfonso Campanile

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maddalena Illario

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge