Network


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

Hotspot


Dive into the research topics where Eudoxia G. Hatzivassiliou is active.

Publication


Featured researches published by Eudoxia G. Hatzivassiliou.


Nature | 2003

CYLD is a deubiquitinating enzyme that negatively regulates NF-κB activation by TNFR family members

Eirini Trompouki; Eudoxia G. Hatzivassiliou; Theodore Tsichritzis; Hannah Farmer; Alan Ashworth; George Mosialos

Familial cylindromatosis is an autosomal dominant predisposition to tumours of skin appendages called cylindromas. Familial cylindromatosis is caused by mutations in a gene encoding the CYLD protein of previously unknown function. Here we show that CYLD is a deubiquitinating enzyme that negatively regulates activation of the transcription factor NF-κB by specific tumour-necrosis factor receptors (TNFRs). Loss of the deubiquitinating activity of CYLD correlates with tumorigenesis. CYLD inhibits activation of NF-κB by the TNFR family members CD40, XEDAR and EDAR in a manner that depends on the deubiquitinating activity of CYLD. Downregulation of CYLD by RNA-mediated interference augments both basal and CD40-mediated activation of NF-κB. The inhibition of NF-κB activation by CYLD is mediated, at least in part, by the deubiquitination and inactivation of TNFR-associated factor 2 (TRAF2) and, to a lesser extent, TRAF6. These results indicate that CYLD is a negative regulator of the cytokine-mediated activation of NF-κB that is required for appropriate cellular homeostasis of skin appendages.


Cellular Signalling | 2011

Mutational analysis of TRAF6 reveals a conserved functional role of the RING dimerization interface and a potentially necessary but insufficient role of RING-dependent TRAF6 polyubiquitination towards NF-κB activation.

Charilaos Megas; Eudoxia G. Hatzivassiliou; Qian Yin; Elli Marinopoulou; Paul Hadweh; Dario A. A. Vignali; George Mosialos

TRAF6 is an E3 ubiquitin ligase that plays a pivotal role in the activation of NF-κB by innate and adaptive immunity stimuli. TRAF6 consists of a highly conserved carboxyl terminal TRAF-C domain which is preceded by a coiled coil domain and an amino terminal region that contains a RING domain and a series of putative zinc-finger motifs. The TRAF-C domain contributes to TRAF6 oligomerization and mediates the interaction of TRAF6 with upstream signaling molecules whereas the RING domain comprises the core of the ubiquitin ligase catalytic domain. In order to identify structural elements that are important for TRAF6-induced NF-κB activation, mutational analysis of the TRAF-C and RING domains was performed. Alterations of highly conserved residues of the TRAF-C domain of TRAF6 did not affect significantly the ability of the protein to activate NF-κB. On the other hand a number of functionally important residues (L77, Q82, R88, F118, N121 and E126) for the activation of NF-κB were identified within the RING domain of TRAF6. Interestingly, several homologues of these residues in TRAF2 were shown to have a conserved functional role in TRAF2-induced NF-κB activation and lie at the dimerization interface of the RING domain. Finally, whereas alteration of Q82, R88 and F118 compromised both the K63-linked polyubiquitination of TRAF6 and its ability to activate NF-κB, alteration of L77, N121 and E126 diminished the NF-κB activating function of TRAF6 without affecting TRAF6 K63-linked polyubiquitination. Our results support a conserved functional role of the TRAF RING domain dimerization interface and a potentially necessary but insufficient role for RING-dependent TRAF6 K63-linked polyubiquitination towards NF-κB activation in cells.


Cellular Signalling | 2014

The PP4R1 subunit of protein phosphatase PP4 targets TRAF2 and TRAF6 to mediate inhibition of NF-κB activation.

Paul Hadweh; Hasem Habelhah; Elliott Kieff; George Mosialos; Eudoxia G. Hatzivassiliou

TRAFs constitute a family of proteins that have been implicated in signal transduction by immunomodulatory cellular receptors and viral proteins. TRAF2 and TRAF6 have an E3-ubiquitin ligase activity, which is dependent on the integrity of their RING finger domain and it has been associated with their ability to activate the NF-κB and AP1 signaling pathways. A yeast two-hybrid screen with TRAF2 as bait, identified the regulatory subunit PP4R1 of protein phosphatase PP4 as a TRAF2-interacting protein. The interaction of TRAF2 with PP4R1 depended on the integrity of the RING finger domain of TRAF2. PP4R1 could interact also with the TRAF2-related factor TRAF6 in a RING domain-dependent manner. Exogenous expression of PP4R1 inhibited NF-κB activation by TRAF2, TRAF6, TNF and the Epstein-Barr virus oncoprotein LMP1. In addition, expression of PP4R1 downregulated IL8 induction by LMP1, whereas downregulation of PP4R1 by RNA interference enhanced the induction of IL8 by LMP1 and TNF. PP4R1 could mediate the dephosphorylation of TRAF2 Ser11, which has been previously implicated in TRAF2-mediated activation of NF-κB. Finally, PP4R1 could inhibit TRAF6 polyubiquitination, suggesting an interference with the E3 ubiquitin ligase activity of TRAF6. Taken together, our data identify a novel mechanism of NF-κB pathway inhibition which is mediated by PP4R1-dependent targeting of specific TRAF molecules.


Cellular Oncology | 2016

Inactivation of CYLD in intestinal epithelial cells exacerbates colitis-associated colorectal carcinogenesis - a short report

Demetrios N. Karatzas; Konstantinos Xanthopoulos; Panorea Kotantaki; Athanasios Pseftogas; Konstantinos Teliousis; Eudoxia G. Hatzivassiliou; Dimitris L. Kontoyiannis; Theofilos Poutahidis; George Mosialos

PurposeCYLD is a tumor suppressor that has been linked to the development of various human malignancies, including colon cancer. The tumor-suppressing function of CYLD is associated with its deubiquitinating activity, which maps to the carboxyl-terminal region of the protein. In the present study we evaluated the role of intestinal epithelial CYLD in colitis-associated cancer using a conditional mouse CYLD inactivation model.MethodsIn order to evaluate the role of CYLD in intestinal epithelial carcinogenesis, mice (IEC-CyldΔ9 mice) that carry a mutation that eliminates the deubiquitinating domain of CYLD in intestinal epithelial cells (IEC) were generated by crossing Villin-Cre transgenic mice to previously generated mice carrying a loxP-flanked Cyld exon 9 (Cyldflx9 mice).ResultsWe found that IEC-CyldΔ9 mice did not present spontaneous intestinal abnormalities up to one year of age. However, upon challenge with a combination of genotoxic (AOM) and pro-inflammatory (DSS) agents we found that the number of adenomas in the IEC-CyldΔ9 mice was dramatically increased compared to the control mice. Inactivation of CYLD in intestinal epithelial cells did not affect the classical nuclear factor-kappaB (NF-κB) and c-Jun kinase (JNK) activation pathways under physiological conditions, suggesting that these pathways do not predispose CYLD-deficient intestinal epithelia to colorectal cancer development before the onset of genotoxic and/or pro-inflammatory stress.ConclusionsOur findings underscore a critical tumor-suppressing role for functional intestinal epithelial CYLD in colitis-associated carcinogenesis. CYLD expression and its associated pathways in intestinal tumors may be exploited for future prognostic and therapeutic purposes.


Journal of Virology | 2005

Induction of Apoptosis by Rewiring the Signal Transduction of Epstein-Barr Virus Oncoprotein LMP1 toward Caspase Activation

Eudoxia G. Hatzivassiliou; Theodore Tsichritzis; George Mosialos

ABSTRACT The Epstein-Barr virus latent membrane protein 1 (LMP1) is an oncoprotein which mimics activated tumor necrosis factor receptor family members. Here we demonstrate the principle that an inducible association of the LMP1 cytoplasmic carboxyl terminus with caspase-8 by a heterodimerizing agent causes apoptosis. This process depends on the catalytic activity of caspase-8 and the ability of LMP1 to oligomerize constitutively at the plasma membrane. Our data indicate that chemical inducers of the association of the LMP1 carboxyl terminus with caspase-8 can kill LMP1-expressing cells selectively. Such compounds could be used as chemotherapeutic agents for LMP1-associated malignancies.


PLOS ONE | 2018

Functional analysis of the C. elegans cyld-1 gene reveals extensive similarity with its human homolog

Paul Hadweh; Iro Chaitoglou; Maria Joao Gravato-Nobre; Petros Ligoxygakis; George Mosialos; Eudoxia G. Hatzivassiliou

The human cylindromatosis tumor suppressor (HsCyld) has attracted extensive attention due to its association with the development of multiple types of cancer. HsCyld encodes a deubiquitinating enzyme (HsCYLD) with a broad range of functions that include the regulation of several cell growth, differentiation and death pathways. HsCyld is an evolutionarily conserved gene. Homologs of HsCyld have been identified in simple model organisms such as Drosophila melanogaster and Caenorhabditis elegans (C. elegans) which offer extensive possibilities for functional analyses. In the present report we have investigated and compared the functional properties of HsCYLD and its C. elegans homolog (CeCYLD). As expected from the mammalian CYLD expression pattern, the CeCyld promoter is active in multiple tissues with certain gastrointestinal epithelia and neuronal cells showing the most prominent activity. CeCYLD is a functional deubiquitinating enzyme with similar specificity to HsCYLD towards K63- and M1-linked polyubiquiting chains. CeCYLD was capable of suppressing the TRAF2-mediated activation of NF-kappaB and AP1 similarly to HsCYLD. Finally, CeCYLD could suppress the induction of TNF-dependent gene expression in mammalian cells similarly to HsCYLD. Our results demonstrate extensively overlapping functions between the HsCYLD and CeCYLD, which establish the C. elegans protein as a valuable model for the elucidation of the complex activity of the human tumor suppressor protein.


Proceedings of the National Academy of Sciences of the United States of America | 1998

Epstein–Barr virus-transforming protein latent infection membrane protein 1 activates transcription factor NF-κB through a pathway that includes the NF-κB-inducing kinase and the IκB kinases IKKα and IKKβ

Bakary S. Sylla; Siu Chun Hung; David Davidson; Eudoxia G. Hatzivassiliou; Nikolai Malinin; David Wallach; Thomas D. Gilmore; Elliott Kieff; George Mosialos


Frontiers in Bioscience | 2002

Cellular signaling pathways engaged by the Epstein-Barr virus transforming protein LMP1.

Eudoxia G. Hatzivassiliou; George Mosialos


Cellular Signalling | 2006

Human ubiquitin specific protease 31 is a deubiquitinating enzyme implicated in activation of nuclear factor-κB

Christos Tzimas; Gianna Michailidou; Minas Arsenakis; Elliott Kieff; George Mosialos; Eudoxia G. Hatzivassiliou


Leukemia Research | 2007

Constitutive CD40 signaling phenocopies the transforming function of the Epstein-Barr virus oncoprotein LMP1 in vitro

Eudoxia G. Hatzivassiliou; Elliott Kieff; George Mosialos

Collaboration


Dive into the Eudoxia G. Hatzivassiliou's collaboration.

Top Co-Authors

Avatar

George Mosialos

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul Hadweh

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Athanasios Pseftogas

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Charilaos Megas

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Christos Tzimas

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Demetrios N. Karatzas

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Elli Marinopoulou

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Gianna Michailidou

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Konstantinos Teliousis

Aristotle University of Thessaloniki

View shared research outputs
Researchain Logo
Decentralizing Knowledge