Network


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

Hotspot


Dive into the research topics where Vassilis G. Gorgoulis is active.

Publication


Featured researches published by Vassilis G. Gorgoulis.


Nature | 2005

Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions

Vassilis G. Gorgoulis; Leandros-Vassilios F. Vassiliou; Panagiotis Karakaidos; Panayotis Zacharatos; Athanassios Kotsinas; Triantafillos Liloglou; Monica Venere; Richard A. DiTullio; Nikolaos G. Kastrinakis; Brynn Levy; Dimitris Kletsas; Akihiro Yoneta; Meenhard Herlyn; Christos Kittas; Thanos D. Halazonetis

DNA damage checkpoint genes, such as p53, are frequently mutated in human cancer, but the selective pressure for their inactivation remains elusive. We analysed a panel of human lung hyperplasias, all of which retained wild-type p53 genes and had no signs of gross chromosomal instability, and found signs of a DNA damage response, including histone H2AX and Chk2 phosphorylation, p53 accumulation, focal staining of p53 binding protein 1 (53BP1) and apoptosis. Progression to carcinoma was associated with p53 or 53BP1 inactivation and decreased apoptosis. A DNA damage response was also observed in dysplastic nevi and in human skin xenografts, in which hyperplasia was induced by overexpression of growth factors. Both lung and experimentally-induced skin hyperplasias showed allelic imbalance at loci that are prone to DNA double-strand break formation when DNA replication is compromised (common fragile sites). We propose that, from its earliest stages, cancer development is associated with DNA replication stress, which leads to DNA double-strand breaks, genomic instability and selective pressure for p53 mutations.


Nature | 2006

Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints.

Jirina Bartkova; Nousin Rezaei; Michalis Liontos; Panagiotis Karakaidos; Dimitris Kletsas; Natalia Issaeva; Leandros-Vassilios F. Vassiliou; Evangelos Kolettas; Katerina Niforou; Vassilis C. Zoumpourlis; Munenori Takaoka; Hiroshi Nakagawa; Frederic Tort; Kasper Fugger; Fredrik Johansson; Maxwell Sehested; Claus L. Andersen; Lars Dyrskjøt; Torben F. Ørntoft; Jiri Lukas; Christos Kittas; Thomas Helleday; Thanos D. Halazonetis; Jiri Bartek; Vassilis G. Gorgoulis

Recent studies have indicated the existence of tumorigenesis barriers that slow or inhibit the progression of preneoplastic lesions to neoplasia. One such barrier involves DNA replication stress, which leads to activation of the DNA damage checkpoint and thereby to apoptosis or cell cycle arrest, whereas a second barrier is mediated by oncogene-induced senescence. The relationship between these two barriers, if any, has not been elucidated. Here we show that oncogene-induced senescence is associated with signs of DNA replication stress, including prematurely terminated DNA replication forks and DNA double-strand breaks. Inhibiting the DNA double-strand break response kinase ataxia telangiectasia mutated (ATM) suppressed the induction of senescence and in a mouse model led to increased tumour size and invasiveness. Analysis of human precancerous lesions further indicated that DNA damage and senescence markers cosegregate closely. Thus, senescence in human preneoplastic lesions is a manifestation of oncogene-induced DNA replication stress and, together with apoptosis, provides a barrier to malignant progression.


Science | 2008

An oncogene-induced DNA damage model for cancer development.

Thanos D. Halazonetis; Vassilis G. Gorgoulis; Jiri Bartek

Of all types of DNA damage, DNA double-strand breaks (DSBs) pose the greatest challenge to cells. One might have, therefore, anticipated that a sizable number of DNA DSBs would be incompatible with cell proliferation. Yet recent experimental findings suggest that, in both precancerous lesions and cancers, activated oncogenes induce stalling and collapse of DNA replication forks, which in turn leads to formation of DNA DSBs. This continuous formation of DNA DSBs may contribute to the genomic instability that characterizes the vast majority of human cancers. In addition, in precancerous lesions, these DNA DSBs activate p53, which, by inducing apoptosis or senescence, raises a barrier to tumor progression. Breach of this barrier by various mechanisms, most notably by p53 mutations, that impair the DNA damage response pathway allows cancers to develop. Thus, oncogene-induced DNA damage may explain two key features of cancer: genomic instability and the high frequency of p53 mutations.


Nature Reviews Molecular Cell Biology | 2010

Genomic instability — an evolving hallmark of cancer

Simona Negrini; Vassilis G. Gorgoulis; Thanos D. Halazonetis

Genomic instability is a characteristic of most cancers. In hereditary cancers, genomic instability results from mutations in DNA repair genes and drives cancer development, as predicted by the mutator hypothesis. In sporadic (non-hereditary) cancers the molecular basis of genomic instability remains unclear, but recent high-throughput sequencing studies suggest that mutations in DNA repair genes are infrequent before therapy, arguing against the mutator hypothesis for these cancers. Instead, the mutation patterns of the tumour suppressor TP53 (which encodes p53), ataxia telangiectasia mutated (ATM) and cyclin-dependent kinase inhibitor 2A (CDKN2A; which encodes p16INK4A and p14ARF) support the oncogene-induced DNA replication stress model, which attributes genomic instability and TP53 and ATM mutations to oncogene-induced DNA damage.


Nature | 2004

Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks

Yentram Huyen; Omar Zgheib; Richard A. DiTullio; Vassilis G. Gorgoulis; Panayotis Zacharatos; Tom J. Petty; Emily A. Sheston; Hestia S. Mellert; Thanos D. Halazonetis

The mechanisms by which eukaryotic cells sense DNA double-strand breaks (DSBs) in order to initiate checkpoint responses are poorly understood. 53BP1 is a conserved checkpoint protein with properties of a DNA DSB sensor. Here, we solved the structure of the domain of 53BP1 that recruits it to sites of DSBs. This domain consists of two tandem tudor folds with a deep pocket at their interface formed by residues conserved in the budding yeast Rad9 and fission yeast Rhp9/Crb2 orthologues. In vitro, the 53BP1 tandem tudor domain bound histone H3 methylated on Lys 79 using residues that form the walls of the pocket; these residues were also required for recruitment of 53BP1 to DSBs. Suppression of DOT1L, the enzyme that methylates Lys 79 of histone H3, also inhibited recruitment of 53BP1 to DSBs. Because methylation of histone H3 Lys 79 was unaltered in response to DNA damage, we propose that 53BP1 senses DSBs indirectly through changes in higher-order chromatin structure that expose the 53BP1 binding site.


Nature Cell Biology | 2011

Interplay between oncogene-induced DNA damage response and heterochromatin in senescence and cancer

Raffaella Di Micco; Gabriele Sulli; Miryana Dobreva; Michalis Liontos; Oronza A. Botrugno; Gaetano Gargiulo; Roberto Dal Zuffo; Valentina Matti; Giovanni d'Ario; Erica Montani; Ciro Mercurio; William C. Hahn; Vassilis G. Gorgoulis; Saverio Minucci; Fabrizio d'Adda di Fagagna

Two major mechanisms have been causally implicated in the establishment of cellular senescence: the activation of the DNA damage response (DDR) pathway and the formation of senescence-associated heterochromatic foci (SAHF). Here we show that in human fibroblasts resistant to premature p16INK4a induction, SAHF are preferentially formed following oncogene activation but are not detected during replicative cellular senescence or on exposure to a variety of senescence-inducing stimuli. Oncogene-induced SAHF formation depends on DNA replication and ATR (ataxia telangiectasia and Rad3-related). Inactivation of ATM (ataxia telangiectasia mutated) or p53 allows the proliferation of oncogene-expressing cells that retain increased heterochromatin induction. In human cancers, levels of heterochromatin markers are higher than in normal tissues, and are independent of the proliferative index or stage of the tumours. Pharmacological and genetic perturbation of heterochromatin in oncogene-expressing cells increase DDR signalling and lead to apoptosis. In vivo, a histone deacetylase inhibitor (HDACi) causes heterochromatin relaxation, increased DDR, apoptosis and tumour regression. These results indicate that heterochromatin induced by oncogenic stress restrains DDR and suggest that the use of chromatin-modifying drugs in cancer therapies may benefit from the study of chromatin and DDR status of tumours.


Modern Pathology | 2000

High risk HPV types are frequently detected in potentially malignant and malignant oral lesions, but not in normal oral mucosa.

Martha Bouda; Vassilis G. Gorgoulis; Nikos G. Kastrinakis; Athina Giannoudis; Efthymia Tsoli; Despina Danassi-Afentaki; Periklis G. Foukas; Aspasia Kyroudi; George Laskaris; C. Simon Herrington; Christos Kittas

Studies on the involvement of the human papillomavirus (HPV) in initiation and progression of oral neoplasia have generated conflicting results. The observed discrepancy is attributable mainly to the varying sensitivity of the applied methodologies and to epidemiologic factors of the examined patient groups. To evaluate the role of HPV in oral carcinogenesis, we analyzed 53 potentially neoplastic and neoplastic oral lesions consisting of 29 cases of hyperplasia, 5 cases of dysplasia, and 19 cases of squamous cell carcinomas, as well as 16 oral specimens derived from healthy individuals. A highly sensitive nested polymerase chain reaction (PCR) assay was used, along with type-specific PCR, restriction fragment length polymorphism analysis, dot blotting, and nonisotopic in situ hybridization. Nested PCR revealed the presence of HPV DNA in 48 of the 53 (91%) pathologic samples analyzed, whereas none (0%) of the normal specimens was found to be infected. Positivity for HPV was independent of histology and the smoking habits of the analyzed group of patients. At least one “high risk” type, such as HPV 16, 18, and 33, was detected by type-specific PCR in 47 (98%) infected specimens, whereas only 1 (2%) squamous cell carcinoma was solely infected by a “low risk” type (HPV 6). HPV 16 was the prevailing viral type, being present in 71% of infected cases. Single HPV 16 and HPV 18 infections were confirmed by restriction fragment length polymorphism. HPV 58 was detected by dot blotting in three hyperplastic lesions. HPV positivity and genotyping were further confirmed, and the physical status of this virus was evaluated by nonisotopic in situ hybridization. Diffuse and punctate signals, indicative of the episomal and integrative pattern of HPV infection, were observed for low- and high-risk types, respectively. Our findings are suggestive of an early involvement of high-risk HPV types in oral carcinogenesis.


International Journal of Cancer | 2009

Hypomethylation of retrotransposable elements correlates with genomic instability in non-small cell lung cancer

Alexandros Daskalos; Georgios Nikolaidis; George Xinarianos; Paraskevi Savvari; Adrian Cassidy; Roubini Zakopoulou; Athanasios Kotsinas; Vassilis G. Gorgoulis; John K. Field; Triantafillos Liloglou

LINE‐1 and Alu elements are non‐LTR retrotransposons, constituting together over 30% of the human genome and they are frequently hypomethylated in human tumors. A relationship between global hypomethylation and genomic instability has been shown, however, there is little evidence to suggest active role for hypomethylation‐mediated reactivation of retroelements in human cancer. In our study, we examined by Pyrosequencing the methylation levels of LINE‐1 and Alu sequences in 48 primary nonsmall cell carcinomas and their paired adjacent tissues. We demonstrate a significant reduction of the methylation levels of both elements (p = 7.7 × 10−14 and 9.6 × 10−7, respectively). The methylation indices of the 2 elements correlated (p = 0.006), suggesting a possible common mechanism for their methylation maintenance. Genomic instability was measured utilizing 11 fluorescent microsatellite markers located on lung cancer hot‐spot regions such as 3p, 5q 9p, 13q and 17p. Hypomethylation of both transposable elements was associated with increased genomic instability (LINE, p = 7.1 × 10−5; Alu, p = 0.008). The reduction of the methylation index of LINE‐1 and Alu following treatment of 3 lung cell lines with 5‐aza‐2′‐deoxycitidine, consistently resulted in increased expression of both elements. Our study demonstrates the strong link between hypomethylation of transposable elements with genomic instability in non‐small cell lung cancer and provides early evidence for a potential active role of these elements in lung neoplasia. As demethylating agents are now entering lung cancer trials, it is imperative to gain a greater insight into the potential reactivation of silent retrotransposons in order to advance for the clinical utilization of epigenetics in cancer therapy.


Cancer Cell | 2013

Mutant p53 Prolongs NF-κB Activation and Promotes Chronic Inflammation and Inflammation-Associated Colorectal Cancer

Tomer Cooks; Ioannis S. Pateras; Ohad Tarcic; Hilla Solomon; Aaron J. Schetter; Sylvia Wilder; Guillermina Lozano; Eli Pikarsky; Tim Forshew; Nitzan Rozenfeld; Noam Harpaz; Steven H. Itzkowitz; Curtis C. Harris; Varda Rotter; Vassilis G. Gorgoulis; Moshe Oren

The tumor suppressor p53 is frequently mutated in human cancer. Common mutant p53 (mutp53) isoforms can actively promote cancer through gain-of-function (GOF) mechanisms. We report that mutp53 prolongs TNF-α-induced NF-κB activation in cultured cells and intestinal organoid cultures. Remarkably, when exposed to dextran sulfate sodium, mice harboring a germline p53 mutation develop severe chronic inflammation and persistent tissue damage, and are highly prone to inflammation-associated colon cancer. This mutp53 GOF is manifested by rapid onset of flat dysplastic lesions that progress to invasive carcinoma with mutp53 accumulation and augmented NF-κB activation, faithfully recapitulating features frequently observed in human colitis-associated colorectal cancer (CAC). These findings might explain the early appearance of p53 mutations in human CAC.


American Journal of Pathology | 2004

Overexpression of the Replication Licensing Regulators hCdt1 and hCdc6 Characterizes a Subset of Non-Small-Cell Lung Carcinomas: Synergistic Effect with Mutant p53 on Tumor Growth and Chromosomal Instability—Evidence of E2F-1 Transcriptional Control over hCdt1

Panagiotis Karakaidos; Stavros Taraviras; Leandros V. Vassiliou; Panayotis Zacharatos; Nikolaos G. Kastrinakis; Dionysia Kougiou; Mirsini Kouloukoussa; Hideo Nishitani; Athanasios G. Papavassiliou; Zoi Lygerou; Vassilis G. Gorgoulis

Replication licensing ensures once per cell cycle replication and is essential for genome stability. Overexpression of two key licensing factors, Cdc6 and Cdt1, leads to overreplication and chromosomal instability (CIN) in lower eukaryotes and recently in human cell lines. In this report, we analyzed hCdt1, hCdc6, and hGeminin, the hCdt1 inhibitor expression, in a series of non-small-cell lung carcinomas, and investigated for putative relations with G(1)/S phase regulators, tumor kinetics, and ploidy. This is the first study of these fundamental licensing elements in primary human lung carcinomas. We herein demonstrate elevated levels (more than fourfold) of hCdt1 and hCdc6 in 43% and 50% of neoplasms, respectively, whereas aberrant expression of hGeminin was observed in 49% of cases (underexpression, 12%; overexpression, 37%). hCdt1 expression positively correlated with hCdc6 and E2F-1 levels (P = 0.001 and P = 0.048, respectively). Supportive of the observed link between E2F-1 and hCdt1, we provide evidence that E2F-1 up-regulates the hCdt1 promoter in cultured mammalian cells. Interestingly, hGeminin overexpression was statistically related to increased hCdt1 levels (P = 0.025). Regarding the kinetic and ploidy status of hCdt1- and/or hCdc6-overexpressing tumors, p53-mutant cases exhibited significantly increased tumor growth values (Growth Index; GI) and aneuploidy/CIN compared to those bearing intact p53 (P = 0.008 for GI, P = 0.001 for CIN). The significance of these results was underscored by the fact that the latter parameters were independent of p53 within the hCdt1-hCdc6 normally expressing cases. Cumulatively, the above suggest a synergistic effect between hCdt1-hCdc6 overexpression and mutant-p53 over tumor growth and CIN in non-small-cell lung carcinomas.

Collaboration


Dive into the Vassilis G. Gorgoulis's collaboration.

Top Co-Authors

Avatar

Athanassios Kotsinas

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Christos Kittas

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Ioannis S. Pateras

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Panayotis Zacharatos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Konstantinos Evangelou

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Michalis Liontos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

George Mariatos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Athanasios G. Papavassiliou

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Ioannis P. Trougakos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge