Network


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

Hotspot


Dive into the research topics where Panayotis Zacharatos is active.

Publication


Featured researches published by Panayotis Zacharatos.


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 | 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.


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.


The Journal of Pathology | 2002

Transcription factor E2F‐1 acts as a growth‐promoting factor and is associated with adverse prognosis in non‐small cell lung carcinomas

Vassilis G. Gorgoulis; Panayotis Zacharatos; George Mariatos; Athanassios Kotsinas; Martha Bouda; Dimitris Kletsas; Panayiotis J. Asimacopoulos; Niki J. Agnantis; Christos Kittas; Athanasios G. Papavassiliou

Numerous upstream stimulatory and inhibitory signals converge to the pRb/E2F pathway, which governs cell‐cycle progression, but the information concerning alterations of E2F‐1 in primary malignancies is very limited. Several in vitro studies report that E2F‐1 can act either as an oncoprotein or as a tumour suppressor protein. In view of this dichotomy in its functions and its critical role in cell cycle control, this study examined the following four aspects of E2F‐1 in a panel of 87 non‐small cell lung carcinomas (NSCLCs), previously analysed for defects in the pRb‐p53‐MDM2 network: firstly, the status of E2F‐1 at the protein, mRNA and DNA levels; secondly, its relationship with the kinetic parameters and genomic instability of the tumours; thirdly, its association with the status of its transcriptional co‐activator CBP, downstream target PCNA and main cell cycle regulatory and E2F‐1‐interacting molecules pRb, p53 and MDM2; and fourthly, its impact on clinical outcome. The protein levels of E2F‐1 and its co‐activator CBP were significantly higher in the tumour area than in the corresponding normal epithelium (p<0.001). E2F‐1 overexpression was associated with increased E2F‐1 mRNA levels in 82% of the cases examined. The latter finding, along with the low frequency of E2F‐1 gene amplification observed (9%), suggests that the main mechanism of E2F‐1 protein overexpression in NSCLCs is deregulation at the transcriptional level. Mutational analysis revealed only one sample with asomatic mutation at codon 371 (Glu→Asp) and one carrying a polymorphism at codon 393 (Gly→Ser). Carcinomas with increased E2F‐1 positivity demonstrated a significant increase in their growth indexes (r=0.402, p=0.001) and were associated with adverse prognosis (p=0.033 by Cox regression analysis). The main determinant of the positive association with growth was the parallel increase between E2F‐1 staining and proliferation (r=0.746, p<0.001), whereas apoptosis was not influenced by the status of E2F‐1. Moreover, correlation with the status of the pRb–p53–MDM2 network showed that the cases with aberrant pRb expression displayed significantly higher E2F‐1 indexes (p=0.033), while a similar association was noticed in the group of carcinomas with deregulation of the p53–MDM2 feedback loop. In conclusion, the results suggest that E2F‐1 overexpression may contribute to the development of NSCLCs by promoting proliferation and provide evidence that this role is further enhanced in a genetic background with deregulated pRb–p53–MDM2 circuitry. Copyright


American Journal of Pathology | 1998

Alterations of the p16-pRb Pathway and the Chromosome Locus 9p21–22 in Non-Small-Cell Lung Carcinomas : Relationship with p53 and MDM2 Protein Expression

Vassilis G. Gorgoulis; Panayotis Zacharatos; Athanassios Kotsinas; Triantofillos Liloglou; Aspasia Kyroudi; Marinos Veslemes; Rassidakis An; Thanos D. Halazonetis; John K. Field; Christos Kittas

The p16-pRb and p53-MDM2 pathways represent vital cell cycle checkpoints. Recent studies provide evidence that these pathways are directly linked via MDM2-pRb interaction and p53 suppression of the RB1 gene. In the present study we investigated the alterations of this G1 phase protein network using immunohistochemical and molecular methods in a series of 68 non-small-cell lung carcinomas (NSCLCs) and correlated the findings with clinicopathological features and prognosis of the patients. Aberrant expression (Ab) of p16 and pRb was observed in 33 (49%) and 27 (40%) of the carcinomas, respectively. Analysis of the region that encodes for p16 by deletion mapping, a polymerase chain reaction (PCR)-based methylation assay and PCR single-strand conformation polymorphism (SSCP) analysis revealed that deletions and transcriptional silencing by methylation might represent the main mechanisms of CDKN2/p16ink4a inactivation in NSCLCs. The results of deletion mapping also suggest that other tumor suppressor genes may reside at the 9p21-22 region, which encodes for CDKN2/MTS1/p16ink4a, p14ARF, and MTS2/p15ink4b. In addition, microsatellite instability was observed with a frequency of 16% in the 9p21-22 chromosome area. Overexpression (P) of p53 and MDM2 proteins was found in 39 (58%) and 47 (70%) of the cases, respectively. A highly significant association was observed between p53 overexpression and p53 mutations (P = 0.006). Statistical analysis of the expression patterns of the biologically relevant molecules (p16/pRb, p53/MDM2, MDM2/pRb, and p53/pRb) showed coincident overexpression of p53 and MDM2 (P = 0.04) and that abnormal pRb was correlated with elevated levels of MDM2 (P = 0.013) and p53 (P = 0.01), respectively. We suggest that deregulated expression of these molecules may act synergistically. An important finding of the study was that multiple impairments (three and four molecules affected) of the p16/pRb/p53/MDM2 network occurred in a large proportion (43%) of the carcinomas. This finding in addition to the absence of correlation with clinical stage of the tumors suggests that multiple hits of this network may be a relatively early event in the development of a subset of NSCLCs. The relationship between the factors examined in the present study, clinicopathological features, and survival of the patients did not reveal any significant correlations with the exception of smoking, which was associated with microsatellite alterations (loss of heterozygosity and microsatellite instability) at the 9p21-22 locus (P = 0.04) and the immunophenotypes p53(P)/MDM2(P) (P = 0.04) and p16(Ab)/pRb(Ab)/p53(P)/MDM2(P) (P = 0.03), respectively. We suggest that in a subset of NSCLCs, simultaneous deregulation of the members of this network may represent one way of initiating the oncogenic procedure whereas in other NSCLC subgroups alternative pathways may play this role.


Human Pathology | 1999

Human papilloma virus (HPV) is possibly involved in laryngeal but not in lung carcinogenesis.

Vassilis G. Gorgoulis; Panayotis Zacharatos; Athanassios Kotsinas; Aspasia Kyroudi; Rassidakis An; John Ikonomopoulos; Calipso Barbatis; C. Simon Herrington; Christos Kittas

Data on human papilloma virus (HPV) involvement in preneoplastic and neoplastic lesions of the larynx and lung are limited and conflicting. The presence of HPV was investigated in a series of laryngeal specimens and non-small cell lung carcinomas (NSCLCs). The laryngeal samples (154) comprised 14 cases with hyperplasia without dysplasia, 49 with dysplasia, and 91 squamous cell carcinomas (SqCCs). The NSCLCs included 31 SqCCs, 32 adenocarcinomas, and 5 undifferentiated large cell carcinomas. Furthermore, we examined, for HPV DNA sequences, 14 bronchial metaplastic squamous lesions located next to cancerous areas. We used a sensitive nested polymerase chain reaction assay (NPCR), dot blotting, and in situ hybridization. The findings were correlated with clinicopathologic features of the patients. In the laryngeal specimens, NPCR analysis showed HPV DNA in 20 (13%) of the 154 specimens. Notably, 19 of 20 HPV-positive cases were carcinomas and only one was a mild dysplastic lesion. Typing of the carcinomas showed single HPV 6, 16, 18, and 33 infection in 1 (1.1%), 12 (13.2%), 2 (2.2%), and 1 (1.1%) samples, respectively, and HPV 6/33, 16/33, and 6/18 coinfection in three carcinomas. In situ hybridization findings were in agreement with PCR results, with the exception of two cases in which HPV 18 DNA was detected only by PCR. HPV was more frequently observed in heavy smokers than in patients with low daily cigarette consumption and nonsmokers (P = .03). There was no correlation between virus infection and gender, grade, and lymph node status of the carcinomas. None of the NSCLCs or adjacent metaplastic squamous epithelium contained HPV DNA sequences. The presented data suggest a contributory role of HPV in late stages of laryngeal carcinogenesis, because all premalignant lesions were negative but one. This study does not support a potential role of HPV in the development of NSCLCs.


The EMBO Journal | 2003

p53 activates ICAM-1 (CD54) expression in an NF-κB-independent manner

Vassilis G. Gorgoulis; Panayotis Zacharatos; Athanassios Kotsinas; Dimitris Kletsas; George Mariatos; Vassilis Zoumpourlis; Kevin M. Ryan; Christos Kittas; Athanasios G. Papavassiliou

Intercellular adhesion molecule‐1 (ICAM‐1) is a crucial receptor in the cell–cell interaction, a process central to the reaction to all forms of injury. Its expression is upregulated in response to a variety of inflammatory/immune mediators, including cellular stresses. The NF‐κB signalling pathway is known to be important for activation of ICAM‐1 transcription. Here we demonstrate that ICAM‐1 induction represents a new cellular response to p53 activation and that NF‐κB inhibition does not prevent the effect of p53 on ICAM‐1 expression after DNA damage. Induction of ICAM‐1 is abolished after treatment with the specific p53 inhibitor pifithrin‐α and is abrogated in p53‐deficient cell lines. Furthermore, we map two functional p53‐responsive elements to the introns of the ICAM‐1 gene, and show that they confer inducibility to p53 in a fashion similar to other p53 target genes. These results support an NF‐κB‐independent role for p53 in ICAM‐1 regulation that may link p53 to ICAM‐1 function in various physiological and pathological settings.


Laboratory Investigation | 2005

p53-Dependent ICAM-1 overexpression in senescent human cells identified in atherosclerotic lesions

Vassilis G. Gorgoulis; Harris Pratsinis; Panayotis Zacharatos; Catherine Demoliou; Fragiska Sigala; Panayiotis J. Asimacopoulos; Athanasios G. Papavassiliou; Dimitris Kletsas

Most normal somatic cells enter a state called replicative senescence after a certain number of divisions, characterized by irreversible growth arrest. Moreover, they express a pronounced inflammatory phenotype that could contribute to the aging process and the development of age-related pathologies. Among the molecules involved in the inflammatory response that are overexpressed in senescent cells and aged tissues is intercellular adhesion molecule-1 (ICAM-1). Furthermore, ICAM-1 is overexpressed in atherosclerosis, an age-related, chronic inflammatory disease. We have recently reported that the transcriptional activator p53 can trigger ICAM-1 expression in an nuclear factor-kappa B (NF-κB)-independent manner (Gorgoulis et al, EMBO J. 2003; 22: 1567–1578). As p53 exhibits an increased transcriptional activity in senescent cells, we investigated whether p53 activation is responsible for the senescence-associated ICAM-1 overexpression. To this end, we used two model systems of cellular senescence: (a) human fibroblasts and (b) conditionally immortalized human vascular smooth muscle cells. Here, we present evidence from both cell systems to support a p53-mediated ICAM-1 overexpression in senescent cells that is independent of NF-κB. We also demonstrate in atherosclerotic lesions the presence of cells coexpressing activated p53, ICAM-1, and stained with the senescence-associated β-galactosidase, a biomarker of replicative senescence. Collectively, our data suggest a direct functional link between p53 and ICAM-1 in senescence and age-related disorders.


The Journal of Pathology | 2004

Distinct expression patterns of the transcription factor E2F-1 in relation to tumour growth parameters in common human carcinomas.

Panayotis Zacharatos; Athanassios Kotsinas; Konstantinos Evangelou; Panagiotis Karakaidos; Leandros-V Vassiliou; Nousin Rezaei; Aspasia Kyroudi; Christos Kittas; Eystratios Patsouris; Athanasios G. Papavassiliou; Vassilis G. Gorgoulis

E2F‐1 is a pivotal transcription factor that integrates signals from a variety of G1/S phase regulators and modulates diverse cellular functions, such as DNA synthesis, repair, mitosis, and apoptosis. Its role in cellular proliferation and apoptosis, as depicted from experimental models and limited reports in human malignancies, remains a matter of debate. Recently, in non‐small cell lung cancer, it was observed that E2F‐1 overexpression was associated with tumour growth, implying an ‘oncogenic’ effect. To clarify further the role of E2F‐1 in carcinogenesis, the investigation was expanded in four of the most common human malignancies by examining its expression status and putative impact on tumour kinetics. These issues were addressed by immunohistochemical and molecular means in 52 breast carcinomas, 42 prostate adenocarcinomas, 58 colon adenocarcinomas, and 77 superficial bladder transitional cell carcinomas (TCCs). The following results were found: (i) in breast carcinomas, E2F‐1 expression correlated with proliferation (p < 0.001) and growth index (p = 0.001); (ii) in prostate adenocarcinomas, absence of E2F‐1 was noted, in contrast to its expression in normal and hyperplastic glands; (iii) in colon adenocarcinomas, E2F‐1 expression was inversely related to growth index (p = 0.001), being expressed in lesions with increased apoptosis (p = 0.001) and low proliferation (p < 0.001); and (iv) in superficial TCCs, E2F‐1 expression correlated with proliferation (p = 0.002). Taken together, these results suggest that E2F‐1 has a growth‐promoting effect in breast carcinomas and superficial TCC, whereas the opposite seems to be the case for colon and prostate cancer. To interpret the above findings, the status of the pRb and p53 tumour suppressor pathways, which are known to affect E2F‐1 activity, was further investigated. The results suggest that the actions of E2F‐1 are mainly dependent on the functionality of these pathways. Nevertheless, the data also imply that p53‐independent pathways may play a nodal role in the function of E2F‐1 in colon cancer. Copyright


American Journal of Pathology | 2002

Proliferation, but Not Apoptosis, Is Associated with Distinct β-Catenin Expression Patterns in Non-Small-Cell Lung Carcinomas : Relationship with Adenomatous Polyposis Coli and G1-to S-Phase Cell-Cycle Regulators

Athamassios Kotsinas; Konstantinos Evangelou; Panayotis Zacharatos; Christos Kittas; Vassilis G. Gorgoulis

beta-catenin (beta-cat) is a versatile component of homotypic cell adhesion and signaling. Its subcellular localization and cytoplasmic levels are tightly regulated by the adenomatous polyposis coli (APC) protein. Mutations in beta-cat (exon 3) or APC (MCR) result in beta-cat aberrant overexpression that is associated with its nuclear accumulation and improper gene activation. Data from experimental models have shown that beta-cat overexpression has a multitude of effects on cell-cycle behavior. In many of these aspects its function depends on major G(1) phase regulators. To the best of our knowledge, most of these issues have never been addressed concurrently in tumors. For this reason we investigated in a panel of 92 non-small-cell lung carcinomas, beta-cat and APC expression, and their relationship with cell-cycle kinetics (PI and AI) and ploidy status. Moreover, the above correlations were examined in relation to the main G(1)/S-phase checkpoint regulators. Four beta-cat immunohistochemical expression patterns [membranous (11.1%), membranous-cytoplasmic (54.3%), cytoplasmic (9.9%), cytoplasmic-nuclear (24.7%)] and three APC immunohistochemical expression patterns [cytoplasmic (37.7%), cytoplasmic-nuclear (58%), nuclear (4.3%)] were observed, which were further confirmed by Western blot analysis on subcellular fractions in representative samples. The frequent presence of beta-cat in the cytoplasm is an indication of aberrant expression, whereas membranous and nuclear localization were inversely related. Absence of mutations in beta-cat (exon 3) and APC (MCR) suggest that beta-cat destruction mechanisms may be functional. However, expression analysis revealed attenuated levels for APC, indicating a residual ability to degrade beta-cat. Decreased levels were associated with loss of heterozygosity at the APC region in 24% of the cases suggesting that additional silencing mechanisms may be involved. Interestingly, the 90-kd APC isoform associated with apoptosis, was found to be the predominant isoform in normal and cancerous lung tissues. The most important finding in our study, was the correlation of nuclear beta-cat immunohistochemical localization with increased proliferation, overexpression of E2F1 and MDM2, aberrant p53, and low expression of p27(KIP), providing for the first time in vivo evidence that beta-cat-associated proliferation correlates with release of E2F1 activity and loss of p53- and p27(KIP)-dependent cell-cycle checkpoints. Loss of these checkpoints is accompanied by low levels of APC, which possibly reflects a diminished ability to degrade beta-cat. Taken together our data indicate that cases with nuclear beta-cat immunohistochemical expression represent a subset of non-small-cell lung carcinomas that have gained an increased proliferation advantage in contrast to the other beta-cat immunohistochemical expression profiles.

Collaboration


Dive into the Panayotis Zacharatos's collaboration.

Top Co-Authors

Avatar

Vassilis G. Gorgoulis

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

Athanassios Kotsinas

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Athanasios G. Papavassiliou

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Evangelos Manolis

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

John Ikonomopoulos

Agricultural University of Athens

View shared research outputs
Researchain Logo
Decentralizing Knowledge