Network


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

Hotspot


Dive into the research topics where Christina Befani is active.

Publication


Featured researches published by Christina Befani.


Journal of Molecular Medicine | 2012

Erratum to: Bortezomib represses HIF-1α protein expression and nuclear accumulation by inhibiting both PI3K/Akt/TOR and MAPK pathways in prostate cancer cells

Christina Befani; Panagiotis J. Vlachostergios; Eleana Hatzidaki; Anna Patrikidou; Sophia Bonanou; George Simos; Christos N. Papandreou; Panagiotis Liakos

Bortezomib represents the first proteasome inhibitor (PI) with demonstrated antitumor activity in the clinical setting, particularly for treatment of hematological malignancies. At the preclinical level, its action is shown to be mediated by induction of growth arrest and apoptosis in many tumor types, including androgen-dependent (AD) and androgen-independent (AI) prostate cancer (PCa) cells. Hypoxia-inducible factor-1α (HIF-1α), which is directly involved in tumor growth, is one of the most studied and promising molecular targets for anti-cancer therapy and is often overexpressed in PCa. Bortezomib has been reported to impair tumor growth by also inhibiting HIF-1α. In this study, we investigated the effect of bortezomib on the expression, activity and localization of HIF-1α in LNCaP (AD) and PC3 (AI) PCa cells. First, we show that hypoxic upregulation of HIF-1α protein levels and activity involves both the PI3K/Akt/mTOR and p44/42 MAPK pathways. Second, bortezomib inhibits expression of HIF-1α protein under both normoxic and hypoxic conditions, represses HIF-1 transcriptional activity and attenuates the release of vascular endothelial growth factor. These effects correlate with the ability of bortezomib to cause dephosphorylation of phospho-Akt, phospho-p70S6K, and phospho-S6RP, thus inactivating a pathway known to be required for HIF-1α protein expression at the translational level. Furthermore, bortezomib also abrogates p44/42 MAPK phosphorylation, which results to reduced nuclear translocation of HIF-1α. Taken together, these results suggest that bortezomib inhibits HIF-1α protein synthesis and its nuclear targeting through suppression of PI3K/Akt/mTOR and MAPK pathways, respectively, in both AD and AI PCa cells.


Journal of Cell Science | 2012

Hypoxia causes triglyceride accumulation by HIF-1-mediated stimulation of lipin 1 expression

Ilias Mylonis; Hiroshi Sembongi; Christina Befani; Panagiotis Liakos; Symeon Siniossoglou; George Simos

Summary Adaptation to hypoxia involves hypoxia-inducible transcription factors (HIFs) and requires reprogramming of cellular metabolism that is essential during both physiological and pathological processes. In contrast to the established role of HIF-1 in glucose metabolism, the involvement of HIFs and the molecular mechanisms concerning the effects of hypoxia on lipid metabolism are poorly characterized. Here, we report that exposure of human cells to hypoxia causes accumulation of triglycerides and lipid droplets. This is accompanied by induction of lipin 1, a phosphatidate phosphatase isoform that catalyzes the penultimate step in triglyceride biosynthesis, whereas lipin 2 remains unaffected. Hypoxic upregulation of lipin 1 expression involves predominantly HIF-1, which binds to a single distal hypoxia-responsive element in the lipin 1 gene promoter and causes its activation under low oxygen conditions. Accumulation of hypoxic triglycerides or lipid droplets can be blocked by siRNA-mediated silencing of lipin 1 expression or kaempferol-mediated inhibition of HIF-1. We conclude that direct control of lipin 1 transcription by HIF-1 is an important regulatory feature of lipid metabolism and its adaptation to hypoxia.


Investigational New Drugs | 2013

Bortezomib overcomes MGMT-related resistance of glioblastoma cell lines to temozolomide in a schedule-dependent manner

Panagiotis J. Vlachostergios; Eleana Hatzidaki; Christina Befani; Panagiotis Liakos; Christos N. Papandreou

SummaryDevelopment of drug resistance after standard chemotherapy for glioblastoma multiforme (GBM) with temozolomide (TMZ) is associated with poor prognosis of GBM patients and is at least partially mediated by a direct DNA repair pathway involving O6-methylguanine methyltransferase (MGMT). This enzyme is under post-translational control by a multisubunit proteolytic cellular machinery, the 26S proteasome. Inhibition of the proteasome by bortezomib (BZ), a boronic acid dipeptide already in clinical use for the treatment of myeloma, has been demonstrated to induce growth arrest and apoptosis in GBM cells. In this study we investigated the effect of sequential treatment with BZ and TMZ on cell proliferation-viability and apoptosis of the human T98G and U87 GBM cell lines. We also tested for an effect of treatment on MGMT expression and important upstream regulators of the latter, including nuclear factor kappa B (NFκB), p44/42 mitogen-activated protein kinase (MAPK), p53, signal transducer and activator of transcription 3 (STAT3) and hypoxia-inducible factor 1α (HIF-1α). The sequence of drug administration for maximal cytotoxicity favored BZ prior to TMZ in T98G cells while the opposite was the case for U87 cells. Maximal efficacy was associated with downregulation of MGMT, reduced IκBα-mediated proteasome-dependent nuclear accumulation of NFκB, attenuation of p44/42 MAPK, AKT and STAT3 activation, and stabilization of p53 and inactive HIF-1α. Collectively, these results suggest that proteasome inhibition by BZ overcomes MGMT-mediated GBM chemoresistance, with scheduling of administration being critical for obtaining the maximal tumoricidal effect of combination with TMZ.


International Journal of Urology | 2012

Bortezomib reverses the proliferative and antiapoptotic effect of neuropeptides on prostate cancer cells.

Konstantinos Tsapakidis; Panagiotis J. Vlachostergios; Ioannis A. Voutsadakis; Christina Befani; Anna Patrikidou; Eleana Hatzidaki; Danai D. Daliani; George Moutzouris; Panagiotis Liakos; Christos N. Papandreou

Objectives:  Neuropeptides are important signal initiators in advanced prostate cancer, partially acting through activation of nuclear factor kappa B. Central to nuclear factor kappa B regulation is the ubiquitin‐proteasome system, pharmacological inhibition of which has been proposed as an anticancer strategy. We investigated the putative role of the proteasome inhibitor bortezomib in neuropeptides signaling effects on prostate cancer cells.


Biochimica et Biophysica Acta | 2013

MgcRacGAP, a cytoskeleton regulator, inhibits HIF-1 transcriptional activity by blocking its dimerization

Aggeliki Lyberopoulou; Ilias Mylonis; George Papachristos; Dimitrios Sagris; Alkmini Kalousi; Christina Befani; Panagiotis Liakos; George Simos; Eleni Georgatsou

Hypoxia inducible factor-1 (HIF-1), a dimeric transcription factor of the bHLH-PAS family, is comprised of HIF-1α, which is inducible by hypoxia and ARNT or HIF-1β, which is constitutively expressed. HIF-1 is involved in cellular homeostasis under hypoxia, in development and in several diseases affected by oxygen availability, particularly cancer. Since its expression is positively correlated with poor outcome prognosis for cancer patients, HIF-1 is a target for pharmaceutical therapy. We have previously shown that male germ cell Rac GTPase activating protein (MgcRacGAP), a regulator of Rho proteins which are principally involved in cytoskeletal organization, binds to HIF-1α and inhibits its transcriptional activity. In this work, we have explored the mechanism of the MgcRacGAP-mediated HIF-1 inactivation. We show that the Myo domain of MgcRacGAP, which is both necessary and sufficient for HIF-1 repression, binds to the PAS-B domain of HIF-1α. Furthermore MgcRacGAP competes with ARNT for binding to the HIF-1α PAS-B domain, as shown by in vitro binding pull down assays. In mammalian cells, ARNT overexpression can overcome the MgcRacGAP-mediated inhibition and MgcRacGAP binding to HIF-1α in vivo inhibits its dimerization with ARNT. We additionally present results indicating that MgcRacGAP binding to HIF-1α is specific, since it does not affect the transcriptional activity of HIF-2, a close evolutionary relative of HIF-1 also involved in hypoxia regulation and cancer. Our results reveal a new mechanism for HIF-1 transcriptional activity regulation, suggest a novel hypoxia-cytoskeleton link and provide new tools for selective HIF-1 inhibition.


Cell Biology International | 2017

Hypoxia upregulates integrin gene expression in microvascular endothelial cells and promotes their migration and capillary‐like tube formation

Christina Befani; Panagiotis Liakos

Tissue hypoxia affects gene expression through the hypoxia‐inducible transcription factors, HIF‐1 and HIF‐2, in both physiological and pathological angiogenesis. Angiogenesis is a complex response of endothelial cells integrating cell proliferation, migration, tube formation, and their interaction with the extracellular matrix through integrin receptors. In this report, we studied the effect of hypoxia on the angiogenic functions of human microvascular endothelial cells (HMEC‐1) as well as on expression of the angiogenic integrins ανβ3, ανβ5, and α5β1. Exposure of HMEC‐1 to hypoxia (1% O2) or to DMOG, a prolyl‐4‐hydroxylase inhibitor, caused significant reduction to their proliferation rate, whereas their migration ability toward laminin‐1 or collagen IV and capillary‐like tube formation were significantly enhanced. In addition, αv, β1, β3, and β5 integrins expression was increased under hypoxia in HMEC‐1, while α5 integrin was not affected. Both HIF‐1 and HIF‐2 protein expression and transcriptional activity were induced under hypoxia in HMEC‐1. The knockdown of either HIF‐1α or HIF‐2α inhibited integrin β3 hypoxic stimulation, suggesting a HIF‐dependent induction of β3 integrin in HMEC‐1. Taken together, our results indicate that hypoxia transcriptionally up‐regulates angiogenic integrins in microvascular endothelial cells along with promoting migration and tube formation of HMEC‐1.


Journal of Cell Science | 2016

HIF-2α phosphorylation by CK1δ promotes erythropoietin secretion in liver cancer cells under hypoxia

Evanthia Pangou; Christina Befani; Ilias Mylonis; Martina Samiotaki; George Panayotou; George Simos; Panagiotis Liakos

ABSTRACT Hypoxia inducible factor 2 (HIF-2) is a transcriptional activator implicated in the cellular response to hypoxia. Regulation of its inducible subunit, HIF-2α (also known as EPAS1), involves post-translational modifications. Here, we demonstrate that casein kinase 1δ (CK1δ; also known as CSNK1D) phosphorylates HIF-2α at Ser383 and Thr528 in vitro. We found that disruption of these phosphorylation sites, and silencing or chemical inhibition of CK1δ, reduced the expression of HIF-2 target genes and the secretion of erythropoietin (EPO) in two hepatic cancer cell lines, Huh7 and HepG2, without affecting the levels of HIF-2α protein expression. Furthermore, when CK1δ-dependent phosphorylation of HIF-2α was inhibited, we observed substantial cytoplasmic mislocalization of HIF-2α, which was reversed upon the addition of the nuclear protein export inhibitor leptomycin B. Taken together, these data suggest that CK1δ enhances EPO secretion from liver cancer cells under hypoxia by modifying HIF-2α and promoting its nuclear accumulation. This modification represents a new mechanism of HIF-2 regulation that might allow HIF isoforms to undertake differing functions. Summary: Phosphorylation of hypoxia inducible factor 2α mediated by casein kinase 1δ increases erythropoietin production by impairing a CRM1-dependent nuclear export mechanism in liver cancer cells.


Journal of Cellular Physiology | 2018

The role of hypoxia-inducible factor-2 alpha in angiogenesis: BEFANI and LIAKOS

Christina Befani; Panagiotis Liakos

Angiogenesis is a key enabling feature of mammalian embryonic development and tumor progression, which provides oxygen and nutrients that are required for vessel growth and tumor cell growth, respectively. Hypoxia is a driver of this phenomenon and is considered to be one of the most potent initiators of angiogenesis both in vitro and in vivo through stabilization of the transcription factors, hypoxia‐inducible factor‐1 and ‐2 (HIF‐1 and HIF‐2). Although these proteins are highly homologous, emerging evidence suggests that they have unique transcriptional targets and differential impact on angiogenesis. Although HIF‐1α is the best known and widely described isoform, recent studies suggest that HIF‐2α is a critical regulator of physiological and pathophysiological angiogenesis and, at least, the similiarly important as HIF‐1α. Indeed, HIF‐2α has been shown to regulate multiple aspects of angiogenesis, including cell proliferation, migration, maturation of blood vessels, and metastasis. In this review, we focus on recent insights into HIF‐2α expression, activation, and function under hypoxic and nonhypoxic conditions. We also summarize the current knowledge on the crosstalk between HIF‐2 and angiogenesis, describing reported phenotypical changes of HIF‐2α genetic models and HIF‐2 target genes implicated in angiogenesis. Finally, we provide a survey of recent pharmacologic strategies to specifically target HIF‐2 activity.


International Journal of Pediatric Otorhinolaryngology | 2018

Serum leukotriene B4 levels, tonsillar hypertrophy and sleep-disordered breathing in childhood

Emmanouel Ι. Alexopoulos; George Haritos; Christina Befani; Georgia Malakasioti; Vassilis Lachanas; Panagiotis Liakos; Konstantinos Gourgoulianis; Athanasios G. Kaditis

OBJECTIVES In children with snoring, increased production of leukotriene B4 (LTB4) may promote tonsillar hypertrophy and sleep-disordered breathing (SDB) or conversely SDB may enhance LTB4 synthesis. We explored whether: i) high LTB4 serum levels predict tonsillar hypertrophy; and ii) SDB severity correlates with LTB4 serum concentration. METHODS Normal-weight children with SDB or controls underwent polysomnography and measurement of LTB4 serum concentration. Tonsillar hypertrophy was the main outcome measure and high LTB4 serum level (>75 t h percentile value in controls) was the primary explanatory variable. Odds ratio (OR) and the corresponding 95% confidence intervals (CI) for tonsillar hypertrophy in children with versus without high LTB4 level were calculated. The control subgroup and subgroups of subjects with increasing SDB severity were compared regarding LTB4 concentration by Kruskal-Wallis test. Spearmans correlation co-efficient was applied to assess the association of LTB4 concentration with SDB severity. RESULTS A total of 104 children with SDB and mean obstructive apnea-hypopnea index-AHI of 4.8 ± 5.3 episodes/h (primary snoring: n = 19; mild SDB: n = 49; moderate/severe SDB: n = 36) and 13 controls (no snoring; AHI: 0.4 ± 0.2 episodes/h) were recruited. The four study subgroups were similar regarding LTB4 serum concentration (P = 0.64). High LTB4 (>170.3 pg/mL) was a significant predictor of tonsillar hypertrophy after adjustment for age and gender (OR 3.0 [1.2-7.2]; P = 0.01). There was no association between AHI or desaturation index and LTB4 serum concentration (r = -0.08; P = 0.37 and r = -0.1; P = 0.30, respectively). CONCLUSION No association was identified between SDB severity and LTB4 levels, but high LTB4 concentration predicted tonsillar hypertrophy.


Sleep and Breathing | 2012

Oxidative stress and inflammatory markers in the exhaled breath condensate of children with OSA

Georgia Malakasioti; Emmanouel I. Alexopoulos; Christina Befani; Kalliopi Tanou; Vasiliki Varlami; Dimitrios C. Ziogas; Panayiotis Liakos; Konstantinos Gourgoulianis; Athanasios G. Kaditis

Collaboration


Dive into the Christina Befani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Athanasios G. Kaditis

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge