@article{DjuzenovaFiedlerMemmeletal.2019, author = {Djuzenova, Cholpon S. and Fiedler, Vanessa and Memmel, Simon and Katzer, Astrid and Sisario, Dmitri and Brosch, Philippa K. and G{\"o}hrung, Alexander and Frister, Svenja and Zimmermann, Heiko and Flentje, Michael and Sukhorukov, Vladimir L.}, title = {Differential effects of the Akt inhibitor MK-2206 on migration and radiation sensitivity of glioblastoma cells}, series = {BMC Cancer}, volume = {19}, journal = {BMC Cancer}, doi = {10.1186/s12885-019-5517-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-200290}, pages = {299}, year = {2019}, abstract = {Background Most tumor cells show aberrantly activated Akt which leads to increased cell survival and resistance to cancer radiotherapy. Therefore, targeting Akt can be a promising strategy for radiosensitization. Here, we explore the impact of the Akt inhibitor MK-2206 alone and in combination with the dual PI3K and mTOR inhibitor PI-103 on the radiation sensitivity of glioblastoma cells. In addition, we examine migration of drug-treated cells. Methods Using single-cell tracking and wound healing migration tests, colony-forming assay, Western blotting, flow cytometry and electrorotation we examined the effects of MK-2206 and PI-103 and/or irradiation on the migration, radiation sensitivity, expression of several marker proteins, DNA damage, cell cycle progression and the plasma membrane properties in two glioblastoma (DK-MG and SNB19) cell lines, previously shown to differ markedly in their migratory behavior and response to PI3K/mTOR inhibition. Results We found that MK-2206 strongly reduces the migration of DK-MG but only moderately reduces the migration of SNB19 cells. Surprisingly, MK-2206 did not cause radiosensitization, but even increased colony-forming ability after irradiation. Moreover, MK-2206 did not enhance the radiosensitizing effect of PI-103. The results appear to contradict the strong depletion of p-Akt in MK-2206-treated cells. Possible reasons for the radioresistance of MK-2206-treated cells could be unaltered or in case of SNB19 cells even increased levels of p-mTOR and p-S6, as compared to the reduced expression of these proteins in PI-103-treated samples. We also found that MK-2206 did not enhance IR-induced DNA damage, neither did it cause cell cycle distortion, nor apoptosis nor excessive autophagy. Conclusions Our study provides proof that MK-2206 can effectively inhibit the expression of Akt in two glioblastoma cell lines. However, due to an aberrant activation of mTOR in response to Akt inhibition in PTEN mutated cells, the therapeutic window needs to be carefully defined, or a combination of Akt and mTOR inhibitors should be considered.}, language = {en} } @article{DjuzenovaFischerKatzeretal.2021, author = {Djuzenova, Cholpon S. and Fischer, Thomas and Katzer, Astrid and Sisario, Dmitri and Korsa, Tessa and Streussloff, Gudrun and Sukhorukov, Vladimir L. and Flentje, Michael}, title = {Opposite effects of the triple target (DNA-PK/PI3K/mTOR) inhibitor PI-103 on the radiation sensitivity of glioblastoma cell lines proficient and deficient in DNA-PKcs}, series = {BMC Cancer}, volume = {21}, journal = {BMC Cancer}, doi = {10.1186/s12885-021-08930-1}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-265826}, year = {2021}, abstract = {Background: Radiotherapy is routinely used to combat glioblastoma (GBM). However, the treatment efficacy is often limited by the radioresistance of GBM cells. Methods: Two GBM lines MO59K and MO59J, differing in intrinsic radiosensitivity and mutational status of DNA-PK and ATM, were analyzed regarding their response to DNA-PK/PI3K/mTOR inhibition by PI-103 in combination with radiation. To this end we assessed colony-forming ability, induction and repair of DNA damage by gamma H2AX and 53BP1, expression of marker proteins, including those belonging to NHEJ and HR repair pathways, degree of apoptosis, autophagy, and cell cycle alterations. Results: We found that PI-103 radiosensitized MO59K cells but, surprisingly, it induced radiation resistance in MO59J cells. Treatment of MO59K cells with PI-103 lead to protraction of the DNA damage repair as compared to drug-free irradiated cells. In PI-103-treated and irradiated MO59J cells the foci numbers of both proteins was higher than in the drug-free samples, but a large portion of DNA damage was quickly repaired. Another cell line-specific difference includes diminished expression of p53 in MO59J cells, which was further reduced by PI-103. Additionally, PI-103-treated MO59K cells exhibited an increased expression of the apoptosis marker cleaved PARP and increased subG1 fraction. Moreover, irradiation induced a strong G2 arrest in MO59J cells (similar to 80\% vs. similar to 50\% in MO59K), which was, however, partially reduced in the presence of PI-103. In contrast, treatment with PI-103 increased the G2 fraction in irradiated MO59K cells. Conclusions: The triple-target inhibitor PI-103 exerted radiosensitization on MO59K cells, but, unexpectedly, caused radioresistance in the MO59J line, lacking DNA-PK. The difference is most likely due to low expression of the DNA-PK substrate p53 in MO59J cells, which was further reduced by PI-103. This led to less apoptosis as compared to drug-free MO59J cells and enhanced survival via partially abolished cell-cycle arrest. The findings suggest that the lack of DNA-PK-dependent NHEJ in MO59J line might be compensated by DNA-PK independent DSB repair via a yet unknown mechanism.}, language = {en} } @phdthesis{Hanselmann2023, author = {Hanselmann, Steffen}, title = {PRC1 serves as a microtubule-bundling protein and is a potential therapeutic target for lung cancer}, doi = {10.25972/OPUS-26631}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-266314}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Protein regulator of cytokinesis 1 (PRC1) is a microtubule-associated protein with essential roles in mitosis and cytokinesis. Furthermore, the protein is highly expressed in several cancer types which is correlated with aneuploidy and worse patient outcome. In this study it was investigated, whether PRC1 is a potential target for lung cancer as well as its possible nuclear role. Elevated PRC1 expression was cell cycle-dependent with increasing levels from S-phase to G2/M-phase of the cell cycle. Thereby, PRC1 localized at the nucleus during interphase and at the central spindle and midbody during mitosis and cytokinesis. Genome-wide expression profiling by RNA sequencing of ectopically expressed PRC1 resulted in activation of the p53 pathway. A mutant version of PRC1, that is unable to enter the nucleus, induced the same gene sets as wildtype PRC1, suggesting that PRC1 has no nuclear-specific functions in lung cancer cells. Finally, PRC1 overexpression leads to proliferation defects, multi-nucleation, and enlargement of cells which was directly linked to microtubule-bundling within the cytoplasm. For analysis of the requirement of PRC1 in lung cancer, different inducible cell lines were generated to deplete the protein by RNA interference (RNAi) in vitro. PRC1 depletion caused proliferation defects and cytokinesis failures with increased numbers of bi- and multi-nucleated cells compared to non-induced lung cancer cells. Importantly, effects in control cells were less severe as in lung cancer cells. Finally, p53 wildtype lung cancer cells became senescent, whereas p53 mutant cells became apoptotic upon PRC1 depletion. PRC1 is also required for tumorigenesis in vivo, which was shown by using a mouse model for non-small cell lung cancer driven by oncogenic K-RAS and loss of p53. Here, lung tumor area, tumor number, and high-grade tumors were significantly reduced in PRC1 depleted conditions by RNAi. In this study, it is shown that PRC1 serves as a microtubule-bundling protein with essential roles in mitosis and cytokinesis. Expression of the protein needs to be tightly regulated to allow unperturbed proliferation of lung cancer cells. It is suggested that besides phosphorylation of PRC1, the nuclear localization might be a protective mechanism for the cells to prevent perinuclear microtubule-bundling. In conclusion, PRC1 could be a potential target of lung cancer as mono therapy or in combination with a chemotherapeutic agent, like cisplatin, which enhanced the negative effects on proliferation of lung cancer cells in vitro.}, language = {en} } @phdthesis{Kaymak2019, author = {Kaymak, Irem}, title = {Identification of metabolic liabilities in 3D models of cancer}, doi = {10.25972/OPUS-18154}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-181544}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Inefficient vascularisation of solid tumours leads to the formation of oxygen and nutrient gradients. In order to mimic this specific feature of the tumour microenvironment, a multicellular tumour spheroid (SPH) culture system was used. These experiments were implemented in p53 isogenic colon cancer cell lines (HCT116 p53 +/+ and HCT116 p53-/-) since Tp53 has important regulatory functions in tumour metabolism. First, the characteristics of the cells cultured as monolayers and as spheroids were investigated by using RNA sequencing and metabolomics to compare gene expression and metabolic features of cells grown in different conditions. This analysis showed that certain features of gene expression found in tumours are also present in spheroids but not in monolayer cultures, including reduced proliferation and induction of hypoxia related genes. Moreover, comparison between the different genotypes revealed that the expression of genes involved in cholesterol homeostasis is induced in p53 deficient cells compared to p53 wild type cells and this difference was only detected in spheroids and tumour samples but not in monolayer cultures. In addition, it was established that loss of p53 leads to the induction of enzymes of the mevalonate pathway via activation of the transcription factor SREBP2, resulting in a metabolic rewiring that supports the generation of ubiquinone (coenzyme Q10). An adequate supply of ubiquinone was essential to support mitochondrial electron transport and pyrimidine biosynthesis in p53 deficient cancer cells under conditions of metabolic stress. Moreover, inhibition of the mevalonate pathway using statins selectively induced oxidative stress and apoptosis in p53 deficient colon cancer cells exposed to oxygen and nutrient deprivation. This was caused by ubiquinone being required for electron transfer by dihydroorotate dehydrogenase, an essential enzyme of the pyrimidine nucleotide biosynthesis pathway. Supplementation with exogenous nucleosides relieved the demand for electron transfer and restored viability of p53 deficient cancer cells under metabolic stress. Moreover, the mevalonate pathway was also essential for the synthesis of ubiquinone for nucleotide biosynthesis to support growth of intestinal tumour organoids. Together, these findings highlight the importance of the mevalonate pathway in cancer cells and provide molecular evidence for an enhanced sensitivity towards the inhibition of mitochondrial electron transfer in tumour-like metabolic environments.}, subject = {Tumor}, language = {en} } @article{KumarNaumannAigneretal.2015, author = {Kumar, Praveen and Naumann, Ulrike and Aigner, Ludwig and Wischhusen, Joerg and Beier, Christoph P and Beier, Dagmar}, title = {Impaired TGF-β induced growth inhibition contributes to the increased proliferation rate of neural stem cells harboring mutant p53}, series = {American Journal of Cancer Research}, volume = {5}, journal = {American Journal of Cancer Research}, number = {11}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-144262}, pages = {3436-3445}, year = {2015}, abstract = {Gliomas have been classified according to their histological properties. However, their respective cells of origin are still unknown. Neural progenitor cells (NPC) from the subventricular zone (SVZ) can initiate tumors in murine models of glioma and are likely cells of origin in the human disease. In both, p53 signaling is often functionally impaired which may contribute to tumor formation. Also, TGF-beta, which under physiological conditions exerts a strong control on the proliferation of NPCs in the SVZ, is a potent mitogen on glioma cells. Here, we approach on the crosstalk between p53 and TGF-beta by loss of function experiments using NPCs derived from p53 mutant mice, as well as pharmacological inhibition of TGF-beta signaling using TGF-beta receptor inhibitors. NPC derived from p53 mutant mice showed increased clonogenicity and more rapid proliferation than their wildtype counterparts. Further, NPC derived from p53\(^{mut/mut}\) mice were insensitive to TGF-beta induced growth arrest. Still, the canonical TGF-beta signaling pathway remained functional in the absence of p53 signaling and expression of key proteins as well as phosphorylation and nuclear translocation of SMAD2 were unaltered. TGF-beta-induced p21 expression could, in contrast, only be detected in p53\(^{wt/wt}\) but not in p53\(^{mut/mut}\) NPC. Conversely, inhibition of TGF-beta signaling using SB431542 increased proliferation of p53\(^{wt/wt}\) but not of p53\(^{mut/mut}\) NPC. In conclusion, our data suggest that the TGF-beta induced growth arrest in NPC depends on functional p53. Mutational inactivation of p53 hence contributes to increased proliferation of NPC and likely to the formation of hyperplasia of the SVZ observed in p53 deficient mice in vivo.}, language = {en} } @article{MainzSarhanRothetal.2022, author = {Mainz, Laura and Sarhan, Mohamed A. F. E. and Roth, Sabine and Sauer, Ursula and Maurus, Katja and Hartmann, Elena M. and Seibert, Helen-Desiree and Rosenwald, Andreas and Diefenbacher, Markus E. and Rosenfeldt, Mathias T.}, title = {Autophagy blockage reduces the incidence of pancreatic ductal adenocarcinoma in the context of mutant Trp53}, series = {Frontiers in Cell and Developmental Biology}, volume = {10}, journal = {Frontiers in Cell and Developmental Biology}, issn = {2296-634X}, doi = {10.3389/fcell.2022.785252}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-266005}, year = {2022}, abstract = {Macroautophagy (hereafter referred to as autophagy) is a homeostatic process that preserves cellular integrity. In mice, autophagy regulates pancreatic ductal adenocarcinoma (PDAC) development in a manner dependent on the status of the tumor suppressor gene Trp53. Studies published so far have investigated the impact of autophagy blockage in tumors arising from Trp53-hemizygous or -homozygous tissue. In contrast, in human PDACs the tumor suppressor gene TP53 is mutated rather than allelically lost, and TP53 mutants retain pathobiological functions that differ from complete allelic loss. In order to better represent the patient situation, we have investigated PDAC development in a well-characterized genetically engineered mouse model (GEMM) of PDAC with mutant Trp53 (Trp53\(^{R172H}\)) and deletion of the essential autophagy gene Atg7. Autophagy blockage reduced PDAC incidence but had no impact on survival time in the subset of animals that formed a tumor. In the absence of Atg7, non-tumor-bearing mice reached a similar age as animals with malignant disease. However, the architecture of autophagy-deficient, tumor-free pancreata was effaced, normal acinar tissue was largely replaced with low-grade pancreatic intraepithelial neoplasias (PanINs) and insulin expressing islet β-cells were reduced. Our data add further complexity to the interplay between Atg7 inhibition and Trp53 status in tumorigenesis.}, language = {en} } @article{PrietoGarciaTomaškovićShahetal.2021, author = {Prieto-Garcia, Cristian and Tomašković, Ines and Shah, Varun Jayeshkumar and Dikic, Ivan and Diefenbacher, Markus}, title = {USP28: oncogene or tumor suppressor? a unifying paradigm for squamous cell carcinoma}, series = {Cells}, volume = {10}, journal = {Cells}, number = {10}, issn = {2073-4409}, doi = {10.3390/cells10102652}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-248409}, year = {2021}, abstract = {Squamous cell carcinomas are therapeutically challenging tumor entities. Low response rates to radiotherapy and chemotherapy are commonly observed in squamous patients and, accordingly, the mortality rate is relatively high compared to other tumor entities. Recently, targeting USP28 has been emerged as a potential alternative to improve the therapeutic response and clinical outcomes of squamous patients. USP28 is a catalytically active deubiquitinase that governs a plethora of biological processes, including cellular proliferation, DNA damage repair, apoptosis and oncogenesis. In squamous cell carcinoma, USP28 is strongly expressed and stabilizes the essential squamous transcription factor ΔNp63, together with important oncogenic factors, such as NOTCH1, c-MYC and c-JUN. It is presumed that USP28 is an oncoprotein; however, recent data suggest that the deubiquitinase also has an antineoplastic effect regulating important tumor suppressor proteins, such as p53 and CHK2. In this review, we discuss: (1) The emerging role of USP28 in cancer. (2) The complexity and mutational landscape of squamous tumors. (3) The genetic alterations and cellular pathways that determine the function of USP28 in squamous cancer. (4) The development and current state of novel USP28 inhibitors.}, language = {en} } @article{ThiemHesbacherKneitzetal.2019, author = {Thiem, Alexander and Hesbacher, Sonja and Kneitz, Hermann and di Primio, Teresa and Heppt, Markus V. and Hermanns, Heike M. and Goebeler, Matthias and Meierjohann, Svenja and Houben, Roland and Schrama, David}, title = {IFN-gamma-induced PD-L1 expression in melanoma depends on p53 expression}, series = {Journal of Experimental \& Clinical Cancer Research}, volume = {38}, journal = {Journal of Experimental \& Clinical Cancer Research}, doi = {10.1186/s13046-019-1403-9}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-201016}, pages = {397}, year = {2019}, abstract = {Background Immune checkpoint inhibition and in particular anti-PD-1 immunotherapy have revolutionized the treatment of advanced melanoma. In this regard, higher tumoral PD-L1 protein (gene name: CD274) expression is associated with better clinical response and increased survival to anti-PD-1 therapy. Moreover, there is increasing evidence that tumor suppressor proteins are involved in immune regulation and are capable of modulating the expression of immune checkpoint proteins. Here, we determined the role of p53 protein (gene name: TP53) in the regulation of PD-L1 expression in melanoma. Methods We analyzed publicly available mRNA and protein expression data from the cancer genome/proteome atlas and performed immunohistochemistry on tumors with known TP53 status. Constitutive and IFN-ɣ-induced PD-L1 expression upon p53 knockdown in wildtype, TP53-mutated or JAK2-overexpressing melanoma cells or in cells, in which p53 was rendered transcriptionally inactive by CRISPR/Cas9, was determined by immunoblot or flow cytometry. Similarly, PD-L1 expression was investigated after overexpression of a transcriptionally-impaired p53 (L22Q, W23S) in TP53-wt or a TP53-knockout melanoma cell line. Immunoblot was applied to analyze the IFN-ɣ signaling pathway. Results For TP53-mutated tumors, an increased CD274 mRNA expression and a higher frequency of PD-L1 positivity was observed. Interestingly, positive correlations of IFNG mRNA and PD-L1 protein in both TP53-wt and -mutated samples and of p53 and PD-L1 protein suggest a non-transcriptional mode of action of p53. Indeed, cell line experiments revealed a diminished IFN-ɣ-induced PD-L1 expression upon p53 knockdown in both wildtype and TP53-mutated melanoma cells, which was not the case when p53 wildtype protein was rendered transcriptionally inactive or by ectopic expression of p53\(^{L22Q,W23S}\), a transcriptionally-impaired variant, in TP53-wt cells. Accordingly, expression of p53\(^{L22Q,W23S}\) in a TP53-knockout melanoma cell line boosted IFN-ɣ-induced PD-L1 expression. The impaired PD-L1-inducibility after p53 knockdown was associated with a reduced JAK2 expression in the cells and was almost abrogated by JAK2 overexpression. Conclusions While having only a small impact on basal PD-L1 expression, both wildtype and mutated p53 play an important positive role for IFN-ɣ-induced PD-L1 expression in melanoma cells by supporting JAK2 expression. Future studies should address, whether p53 expression levels might influence response to anti-PD-1 immunotherapy.}, language = {en} } @phdthesis{Ulrich2012, author = {Ulrich, Tanja}, title = {Function of Lin9 in vivo and MAP3K4-p38 signaling regulates p53 mediated cell cycle arrest after defective mitosis}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-73975}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Eine genaue Kontrolle des Verlaufs durch die Mitose ist entscheidend f{\"u}r die Gew{\"a}hrleistung genomischer Stabilit{\"a}t und f{\"u}r die Vermeidung von Aneuploidy. Der DREAM Komplex ist ein wichtiger Regulator der Expression von mitotischen Genen. Die Depletion der DREAM-Untereinheit Lin9, f{\"u}hrt zu einer verminderten Expression von G2/M Genen und beeintr{\"a}chtigt die Proliferation. In konditionellen knockout Mauszellen (MEFs) verursacht das Ausschalten von Lin9 Defekte in Mitose und Zytokinese und l{\"o}st vorzeitige Seneszenz aus, um eine weitere Zellproliferation zu verhindern. In dieser Arbeit konnte gezeigt werden, dass der seneszente Ph{\"a}notyp in Lin9 knockout MEFs unabh{\"a}ngig von den beiden Tumorsuppressor-Signalwegen p53-p21 und p16-pRB induziert wird. Untersuchungen mit dem konditionellen Lin9 knockout Mausmodell verdeutlichten die wichtige Funktion von Lin9 in der Regulierung der mitotischen Genexpression und der Proliferation in vivo. Das Fehlen von Lin9 f{\"u}hrte zu einer verringerten Proliferation in den Krypten des D{\"u}nndarms und verursachte eine Atrophie des Darmepithels und einen schnell eintretenden Tod der Tiere. Im zweiten Teil der Arbeit wurden Signalwege untersucht, die nach fehlerhafter Zytokinese zu einem p53 vermittelten G1-Arrest f{\"u}hren. Hierf{\"u}r wurde ein chemischer Inhibitor der mitotischen Kinase Aurora B verwendet. Mit Hilfe eines Hochdurchsatz siRNA Screens wurde die MAP Kinase MAP3K4 als Aktivator des p53 Signalwegs identifiziert. Es konnte gezeigt werden, dass MAP3K4 die Stresskinase p38b aktiviert, um den p53 vermittelten Zellzyklusarrest in tetraploiden Zellen auszul{\"o}sen. Dabei wurde p38b nach Hemmung von Aurora B f{\"u}r die transkriptionelle Aktivierung des p53 Zielgens p21 ben{\"o}tigt. Im Gegenteil dazu erfolgte die Phosphorylierung, Stabilisierung und die Rekrutierung von p53 an den p21 Promoter unabh{\"a}ngig von p38. Die teilweise Hemmung von Aurora B zeigte, dass fehlerhafte Segregation von Chromosomen auch den MAP3K4-p38-p53 Signalweg aktiviert und l{\"a}sst darauf schließen, dass subtile Defekte in der Mitose ausreichen diesen Stress-Signalweg zu induzieren. Obwohl p38 f{\"u}r den G1 Zellzyklusarrest nach mitotischen Sch{\"a}den erforderlich war, f{\"u}hrte die gleichzeitige Inhibierung von p38 und Aurora B {\"u}ber einen l{\"a}ngeren Zeitraum zu einer verringerten Proliferation, vermutlich aufgrund verst{\"a}rkter Apoptose. Es ist anzunehmen, dass der MAP3K4-p38-p53 Signalweg generell nach Defekten in der Mitose oder Zytokinese aktiviert wird um Zellen in G1 zu arretieren und um chromosomale Instabilit{\"a}t zu vermeiden.}, subject = {Mitose}, language = {en} } @phdthesis{Vukicevic2004, author = {Vukicevic, Vladimir}, title = {Mechanisms of apoptosis modulation and their contribution to genomic instability in tumor cells}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-10605}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2004}, abstract = {The concept of programmed cell death has been increasingly considered from various aspects since early 1970's. Primarily, knowledge of apoptosis referred to morphological changes in which chromatin is condensed and increasingly fragmented, revealed as small structure in the nucleus. The membrane shrinks and the cell becomes dense as can be seen by flow cytometry. Interestingly, similar modes of cell deletion were observed in nematodes indicating that apoptosis is a highly conserved machinery. Three Caeonorhabditis elegans gene products are found to have high homology with mammalian apoptotic genes: CED-9 inhibits apoptosis and is related to bcl-2; CED-3 and CED-4 promote apoptosis and are related to caspase 9 and APAF-1. Apoptosis is not accidental death, but a highly controlled and medically important molecular process. More general terms such as 'physiological' or 'regulated' cell death cover different morphologies and sequences. Programmed suicide of cells that were subjected to toxic exogenous and endogenous stimuli plays a key role in understanding cancer development and its treatment. Apoptosis involves sequences of events that may overlap and play contradictory or antagonistic roles in cell death. Generally, the ability to trigger apoptotic processes in cancer cells would benefit an organism by keeping homeostasis intact. Programmed cell death is a regularly present mechanism, for instance, in lymphocyte recruitment in the thymus where immature lymphocytes may recognize host antigens. Therefore, such lymphocytes become apoptotic and are removed by macrophages. Removal prevents possible autoimmune diseases. Unlike apoptosis, necrosis is a passive process of cell death recognizable by membrane morphological changes and accompanied by leakage of intracellular material into intercellular space that may cause inflammation in the organism. Signals that may initiate apoptosis are generally classified into two groups: signals that launch extrinsic apoptotic pathways starting with aggregation of death receptors and intrinsic apoptotic pathways starting with disruption of intracellular homeostasis such as the release of mitochondrial factors or DNA degradation. Early in the process, apoptotic signals may lead to a broad range of signaling mechanisms such as DNA repair and assessment of DNA damage (check points). Thus, failure in any of these steps can cause a defective apoptotic response that plays a decisive role in both tumorigenesis and drug resistance in tumor treatment. More distinctly, the capability of cancer cells to go into apoptosis prevents further neoplastic changes. Generally, the purpose of this study is to investigate the balance between formation of genomic damage and induction of apoptosis under genotoxic stress. After genotoxic insult there are different possibilities for the fate of a cell (Figure 1). The genomic integrity is analyzed at cellular checkpoints, usually leading to a delay in cell cycle progression if DNA was damaged. Mutations in genes such as p53 and p21 change the cellular response to genotoxic stress and may alter the balance between apoptosis and genomic damage. However, p53 is usually mutated or not expressed in 70\% of human tumors. Alterations in p53 states that reflect distinct apoptotic response upon induction of DNA damage were examined. In this study, three cell lines with distinct p53 states were used: TK6 harboring wild-type p53, WTK1 with mutated p53 and NH32 with knocked out p53. In the present work we applied different approaches to investigate the correlation between DNA damage and apoptotic responsiveness in cancer cell lines with different p53 states or in hormone responsive cell lines with over expressed bcl-2 gene. We were focused on effects caused by temporary down regulation of the p53 and Bcl-2 activity in human lymphoblastoid cell lines. In addition, we investigated the impact of estradiol-induced proliferation on apoptosis and DNA damage in stably transfected cells with bcl-2gene.}, subject = {Apoptosis}, language = {en} }