• No results found

Distribution of DNAdouble-strand break repairproteins DNA-PK

N/A
N/A
Protected

Academic year: 2022

Share "Distribution of DNAdouble-strand break repairproteins DNA-PK"

Copied!
27
0
0

Loading.... (view fulltext now)

Full text

(1)

UPTEC X 01 005 ISSN 1401-2138 JAN 2001

MALIN NORDING

Distribution of DNA

double-strand break repair proteins DNA-PK CS and Mre11 after exposure to ionizing radiation

Master’s degree project

(2)

Molecular Biotechnology Programme Uppsala University School of Engineering UPTEC X 01 005

Date of issue 2001-01

Author

Malin Nording

Title (English)

Distribution of DNA double-strand break repair proteins DNA- PK

CS

and Mre11 after exposure to ionizing radiation

Title (Swedish) Abstract

DNA is occasionally damaged and efficient repair mechanisms are crucial for the cell. Here the proteins DNA-PKCS and Mre11, involved in the non-homologous end joining process of double-strand break repair, were investigated. Double-strand breaks in DNA were induced with different kinds of radiation. γ-irradiation from a 60Co source was used as an example of low linear energy transfer (LET) radiation and cyclotron accelerated 14N7+ as an example of high LET radiation. The distribution of DNA-PKCS and Mre11 after various times of repair was studied using a method based on immunohistochemistry. The results showed that Mre11 foci formation was affected by LET and dose, and that the DNA-PK inhibitor wortmannin also has an impact on Mre11 foci formation, suggesting activation of Mre11 by DNA-PKCS Further it seems like Mre11 foci sometimes are located in holes without DNA-PKCS. Keywords

DNA-PKCS, Mre11, double-strand break, repair, ionizing radiation, LET, fluorescence Supervisors

Bo Stenerlöw

Division of Biomedical Radiation Sciences, Uppsala University Examiner

Jörgen Carlsson

Division of Biomedical Radiation Sciences, Uppsala University

Project name Sponsors

Language

English

Security

ISSN 1401-2138 Classification

Supplementary bibliographical information Pages

27

Biology Education Centre Biomedical Center Husargatan 3 Uppsala Box 592 S-75124 Uppsala Tel +46 (0)18 4710000 Fax +46 (0)18 555217

(3)

S U M M A R Y IN S W E D I S H

Sammanfattning

Ibland skadas arvsmassan (DNA) i cellerna. För att cellen ska kunna överleva och behålla sina funktioner behövs en mekanism som lagar brotten på DNA. Det är olika proteiner som sköter reparationen och i detta examensarbete undersöks två reparationsproteiner, nämligen de som kallas Mre11 och DNA-PKcs.

För induktionen av brott på DNA används i detta arbete två olika typer av strålning.

Cellerna får olika tider på sig att reparera skadorna och därefter fixeras cellerna, det vill säga dödas på ett sätt som bevarar strukturerna i cellen. Man behandlar cellerna med en första antikropp, som känner igen de intressanta proteinerna, och sedan med en andra antikropp, som känner igen den första antikroppen. Ett självlysande färgämne sitter fast på den andra antikroppen, som gör att Mre11 blir rött och DNA- PKcs blir grönt när man tittar på cellerna i mikroskop. På så sätt kan man undersöka hur proteinerna beter sig efter olika lång tid av reparation.

Resultaten visar att proteinerna till exempel påverkas av vilken typ av strålning de utsätts för.

Malin Nording

Examensarbete 20 p i Molekylär bioteknikprogrammet Uppsala universitet, januari 2001

(4)

C O N T E N T S

ABBREVIATIONS...5

INTRODUCTION...6

Ionizing radiation ... 6

DNA damage and repair... 9

DNA-dependent protein kinase ... 9

Mre11... 11

Aim of the study ... 12

MATERIALS AND METHODS...13

Cells and culture condition ... 13

Irradiation protocols ... 13

γ-irradiation... 14

Exposure to nitrogen ions ... 14

Experimental groups and time points... 14

Immunofluorescent staining... 15

Antibodies... 16

Experimental procedure ... 16

Microscopy ... 17

RESULTS AND DISCUSSION...18

CONCLUSIONS...24

REFERENCES...25

ACKNOWLEDGEMENTS...27

(5)

A B B R E V I A T I O N S

DNA deoxyribonucleic acid

DNA-PK DNA-dependent protein kinase

DSB double-strand break

FCS foetal calf serum

HR homologous recombination

kbp kilo base pair

LET linear energy transfer

Mbp mega base pair

NHEJ non-homologous end joining

PBS phosphate buffer saline

RBE relative biological effectiveness

(6)

I N T R O D U C T I O N

In everyday life the genome in our cells runs the risk of being damaged. A sunbeam hits our skin and transfer its energy into the nucleus of the cells where it may cause breaks in the DNA. The gas radon emits α-particles and can give rise to the same effect. But it is not only radiation that jeopardises the integrity of the genome. Other examples of damaging agents are different kinds of harmful chemicals.

The actual damage of the DNA can be of different kinds, sometimes there is a base damage and other times there is a single or double strand break. The double strand break (DSB) is considered to be the critical one after exposure to ionizing radiation (reviewed by Iliakis, 1991). The damage can be lethal to the cell, mutations can arise, or in worst case scenario there is a possibility that the cell develops into a tumour cell. The tremendous importance of effective cellular DSB repair mechanisms is obvious. This is consequently a very interesting field of research and a lot of details regarding the DSB repair mechanism still lays ahead to be investigated.

In this Degree project the emphasis lies on the investigation of two of the proteins involved in DSB repair, Mre11 and DNA-PKCS. These proteins represent different aspects of the DSB repair mechanism and it is still unclear how they co-operate in the process of DSB repair.

Ionizing radiation

The definition of ionizing radiation states that it is a particle or photon that can interact with a target and subsequently cause ionization. This can take place whenever the energy of the particle or photon exceeds the ionization potential, the energy it takes to remove an electron from an atom or molecule. Two examples of ionization radiation are γ-radiation and X-rays. The reason why these types of electromagnetic radiation have the ability to ionize target molecules is their short wavelength (in the range of the size of an atom) and hence very high frequency.

Because of the short wavelength the energy content is high enough to disrupt biological structures at the atomic or molecular level. Processes that result in ionization are particle interactions such as collisions and photon interactions such as photoelectric effect, Compton effect, and pair production (Nias, 1990).

Ionizing radiation is invisible and humans have no sensory organs to detect it, still it creates a very hazardous existence for the cells in the body. 10 Gy X-rays (1 Gy

(7)

equals an energy deposition of 1 Joule/kg) kill almost all mammalian cells. This amount of energy corresponds to an increase of the temperature by only 0.002 °C if the energy would have been deposited in the form of heat. In the case of ionizing radiation transfer of large amounts of energy from the particles or photons to relatively small volumes in the cell causes more severe effects compare to heat. If the target of the energy transfer happens to be the DNA molecule then there can be disastrous consequences for cell survival and the integrity of the genome. DNA damage produced by ionizing radiation can lead to cell killing, mutagenesis, and cell transformation (reviewed by Ward, 1988).

The energy deposition is measured by the spacing of ionizations along a linear path and is described as linear energy transfer (LET). High LET is caused by densely ionizing radiation while low LET is a result of sparsely ionizing radiation (figure 1).

Figure 1. Schematic illustration of the interaction between radiation with different ionization density (high LET ion and low LET electron) and DNA (by courtesey of J. Carlsson1). Every circle corresponds to one ionization.

The main source of human exposure to high LET radiation in nature is inhaled radon gas (Nias, 1990) which emits α-particles, but it is also possible to produce high LET radiation artificially for instance in a cyclotron. One example of cyclotron produced high LET is 14N7+. The γ-radiation (photons) resulting from 60Co decay is an example of low LET radiation.

1 Lundqvist, H. and Carlsson, J., Nuclide Technique, Internal Compendium (1999)

(8)

To describe the biological effect of ionizing radiation, which depends upon how densely it is deposited, relative biological effectiveness (RBE) is used. Figure 2 illustrates the concept.

Figure 2. DNA double-strand break induction after irradiation with nitrogen ions, LET = 125 keV/µm, and 60Co photons, LET < 0.5 keV/µm (Höglund et al., 2000 ).

The number of DSBs induced per cell versus absorbed dose is plotted for two different kinds of radiation. The nitrogen ions induce more DSBs/cell compare to photons. The photons are used as a reference radiation and the definition states that RBE in this case equals Dγ/DN ions. For example, 52 DSBs/cell are induced when the cells are exposed to 1 Gy nitrogen ions (LET 125 keV/µm), while the cells have to be exposed to 1.5 Gy γ-irradiation before the same amount DSBs are induced. This gives a RBE of 1.5, since 1.5/1 equals 1.5. The RBE for reference low LET photon radiation (X-rays or 60Co photons) is by definition one. High LET radiation creates much more severe effects on the target molecule than low LET does and RBE-values increase with increasing LET. The greater LET the greater RBE with an increasing risk of cell death, mutations, and cell transformations as a result.

A further aspect of ionizing radiation to consider is the direct or indirect effect it can have on its target. In a biological perspective the affect on a molecule is not dependent upon whether the action of the ionizing radiation is direct or indirect and both effects are important. A prerequisite for the indirect effect to take place is the ionization of water when the aqueous free radicals OHand Hare produced. Water is common in human cells and the aqueous free radicals can cause ionizations in an indirect manner when they interact with biological targets such as DNA. A direct effect of ionizing radiation is created when the target molecule is ionized without the participation of radicals. High LET radiation ionizes the target molecule mainly in a direct way while low LET radiation operates mainly in an indirect way (Nias, 1990).

0 250 500 750

0 5 10

Dose (Gy)

DSB/cell

Photons N ions

(9)

DNA damage and repair

Damages on DNA are prevalent and dangerous to the cell. DSBs created by for example ionizing radiation or during the process of DNA replication (reviewed by Haber, 1999) may be the most serious lesion. Every single one of the DSBs is a possible source of mutation and cell death. If the DSBs are repaired improperly, they can even lead to cancer. With this in mind it is astonishing that human cells are able to withstand about ten DSBs caused by replication during every cell cycle (Haber, 1999).

There exist different pathways for the DSB repair, with homologous recombination (HR) and non-homologous end joining (NHEJ) as the outstanding ones (reviewed by Karran, 2000). In mammalian cells NHEJ is the major pathway and involves the proteins DNA ligase IV, XRCC4, DNA-dependent protein kinase (DNA-PK), Rad50, Mre11, NBS1, and SIR2 (reviewed by Critchlow and Jackson, 1998).

The DSB repair shows a biphasic behaviour with a slow and a fast component. The half-time for the fast component is 15 minutes and for the slow component it is 2-3 hours. (Stenerlöw et al., 2000). Further it is shown that the fast component is responsible for the majority of the DSB repair, while the rejoining by the slow component increases with increasing LET. This probably reflects a greater complexity of the breaks in the case of high LET radiation. The strands may for example be torn to pieces in the vicinity of the break leaving mismatching ends in the open. Furthermore, high LET radiation also induces correlated DSBs, non-randomly distributed within 10 kbp - 2 Mbp, that might affect the reparability (Höglund et al., 2000, Stenerlöw et al., 2000).

DNA-dependent protein kinase

DNA-dependent protein kinase (DNA-PK) is an abundant serine-threonine protein kinase in the nucleus of human cells where its general function is to phosphorylate serine and threonine residues in specific proteins when bound to DNA. DNA-PK consists of the heterodimer Ku composed of Ku70 and Ku802, and the catalytic subunit DNA-PKCS. A model presented by DiBiase et al. (2000) suggests that the DNA-PKCS is attached to the nuclear matrix, close to other components of the NHEJ complex. Ku binds to the free DNA ends in the presence of DSBs in the chromatin loop, and is then recruited by DNA-PKCS (figure 3). Subsequently DNA-PKCS

2 Sometimes referred to as Ku86.

(10)

phosphorylates Ku and additional proteins in the NHEJ complex and the DSB is repaired. The model is supported by the fact that Ku is capable of self-association when bound to DNA (Cary, 1997).

Figure 3. A model purposed for DSB repair by the NHEJ mechanism (adopted from DiBiase et al., 2000).

The structure of DNA-PKCS has been determined by electron crystallography (Leuther et al., 1999). The interaction between DNA-PKCS and DNA is proposed to activate the protein kinase in a strand length dependent way. The double stranded DNA may bind to a cavity in the protein kinase at the same time as a short single strand is threaded into an adjacent cavity (figure 4). Processing of the DNA ends leads to the formation of longer unpaired single strands, that will inhibit the protein kinase when they become long enough to penetrate deeply into the cavity. The other single strand is supposed to be threaded into an opening of a DNA-PKCS molecule bound to the opposing DNA end, bringing the two ends together for ligation.

Kinase on Kinase off

Figure 4. A model for binding of single stranded and double stranded DNA to the cavities of DNA- PKCS (adopted from Leuther et al., 1999).

s

Chromatin loop

DNA-PKcs NHEJ Ku

Ku

Nuclear matrix

(11)

DNA-PK has apparently the role of bringing the DNA ends together as well as activating proteins involved in the NHEJ mechanism. The activation of DNA-PKCS by DNA might be through the induction of conformational changes in the enzyme whereas the active site is exposed (Leuther et al., 1999). In the process of covalently joining the strands, the XRCC4 protein and DNA ligase IV are involved (Critchlow et al., 1997) and a nuclease removes unpaired DNA flaps. The DSB ends are fused improperly by NHEJ and when the repair is complete the rejoined DNA has small deletions.

DNA-PK is inhibited in a non-competetive and irreversible manner by the fungal metabolite wortmannin with increased cell radiosensitivity to killing as a result (Boulton et al., 1996). Cells with functional DNA-PKCS treated with wortmannin and cells lacking DNA-PKCS are equivalent radiosensitive to killing (DiBiase et al., 2000).

Mre11

Mre11 is an abundant nuclear protein involved in a variety of processes in the cell (reviewed by Haber, 1998). Together with other proteins it plays a very important role in telomere maintenance, but it is also a participant in DSB repair. The action of Mre11 in DSB repair is somewhat unclear, though it has been proposed to have a nucleolytic role (Paull and Gellert, 1998).

It has been shown that Mre11, Rad50 and NBS1 assemble into a complex (Carney et al., 1998) and when exposed to ionizing radiation Mre11 and Rad50 form nuclear foci (accumulations) that colocalize (Maser et al., 1997). By developing a method for partial irradiation of the cell nucleus it has further been shown that Mre11 foci colocalize with DSBs and that DNA lesions do not move around in the cell nucleus.

Repair proteins such as Mre11 are consequently recruited to the sites of DSBs (Nelms et al., 1998).

It is worth to consider the large amount molecules that have to be present before a focus is detectable when only one set of repair proteins would be theoretically enough to repair one DSB. Difficulty repairing certain breaks is one possible reason why proteins involved in DSB repair form foci (Paull et al., 2000).

(12)

Aim of the study

The purpose with this Master’s Degree project was to establish protocols for immunofluorescent staining of DNA-PKCS and Mre11 and to conduct experiments based on these protocols.

Questions to be answered were:

Is the formation of Mre11 foci affected by LET and dose?

Is it possible that wortmannin, a DNA-PK inhibitor, has any affect on the formation of Mre11 foci?

How are DNA-PKCS and Mre11 correlated to each other?

(13)

M A T E R I A L S A N D M E T H O D S

Cells and culture condition

Low passages of normal human fibroblasts GM5758 (Human Genetic Mutant Cell Repository, Camden, NJ, USA) were used in all of the experiments. These cells are not so specialized and are therefore in many ways similar to other cells in the body.

To insure studies on a homogenous system, the cells were allowed to grow to confluence before any experiments were conducted. Consequently, normal human fibroblasts, grown to confluence with the majority of the cells in the G1/G0 phase of the cell cycle, provide a good model system for studying the DSB repair process.

The cells were grown in Eagle minimal essential medium (MEM) supplemented with 15% foetal calf serum (FCS), 2 mM L-glutamine, 100 µg/ml streptomycin, 100 IU/ml penicillin, 2 × concentration of vitamins and both non-essential, and essential amino acids (all added mixtures obtained from Biochrom KG, Germany). The cells were maintained in cell flasks at 37°C in a humidified atmosphere of 5% CO2 and 95% air.

To be able to analyse the cells they had to be grown in monolayer on microscope slides and a procedure for this purpose was established. The cells in the flask were trypsinized with 0.5 ml trypsin and then 2 ml complete medium was added. About 0.1 ml cell suspension was put as a spot with diameter of about 2 cm on 76x26 mm microscope slides (KEBO-Lab, Sweden), which were kept in petri dishes (Becton Dickinson, Franklin Lakes, NJ, USA) with space for three slides. The cells adhered for one hour before complete medium was added. The cells were grown to confluence before irradiation took place.

Irradiation protocols

Two kinds of radiation were used, γ-radiation from a 60Co source and cyclotron produced 14N7+, i.e. particle radiation. The cells exposed to γ-radiation were either treated with wortmannin (Sigma, Saint Lois, Missouri, USA) or left untreated. The irradiation protocols were conducted at The Svedberg Laboratory, Uppsala University. During the exposure to γ-irradiation all samples were put on ice and during exposure to nitrogen ions the cells were cooled to prevent the action of any repair processes.

(14)

γ-irradiation

Cells were either grown for 50 minutes in complete medium with 20 µM wortmannin at 37˚C or left untreated, i.e. grown in complete medium without wortmannin.

The cells were cooled in cold complete medium and put on ice approximately 15 minutes prior to irradiation. The cells were exposed to γ-radiation from a 60Co source (1.2 MeV photons, LET ~0.5 keV/µm) at a dose rate of 1.6 Gy/min. The cells received a total dose of 10 Gy. After irradiation the cells were left in complete medium to repair breaks in DNA at 37˚C. Cells treated with wortmannin were both cooled and recovered in complete medium with 20 µM wortmannin.

Exposure to nitrogen ions

The cells were cooled in cold serum free medium and put on ice approximately 15 minutes prior to irradiation. The cells were exposed to 14N7+, accelerated in the Gustaf Werner synchrocyclotron (LET 125 keV/µm). The slides were put in the ice-cooled sample holder (figure 5) where the cells received a total dose of 1 or 10 Gy. After irradiation the cells were allowed to recover in complete medium at 37˚C.

Figure 5. Experimental set-up for ion irradiation at the Biomedical Unit, The Svedberg Laboratory, Uppsala University (see Stenerlöw et al., 1996 for details). Gold foil (A), transmission chamber (B), plastic absorber (C), scanning device for Markus chamber (D) and ice-cooled sample holder (E). [By courtesy of T. Hartman, The Svedberg Laboratory, Uppsala, Sweden.]

Experimental groups and time points

Cells exposed to 10 Gy γ-radiation treated with or without wortmannin were allowed to recover for 0, 130, and 240 minutes. During that time the repair mechanism was

(15)

active and the behaviour of Mre11 and DNA-PKCS could be monitored. One additional repair time was allowed for cells left untreated, namely 24 hours.

Unirradiated control cells were treated in the same way as cells allowed to recover without wortmannin for 0 and 240 minutes respectively. In experiments with wortmannin control cells were allowed to recover for 240 minutes. During irradiation the controls were left on ice.

In the case of irradiation with 10 Gy nitrogen ions, the cells were allowed to recover for 0, 130 minutes, and 24 hours. Cells exposed to 1 Gy nitrogen ions were allowed to recover for 130 minutes.

Unirradiated control cells were treated in the same way as cells exposed to 10 Gy nitrogen ions and allowed to recover for 130 minutes and 24 hours respectively.

During irradiation the controls were left on ice.

Immunofluorescent staining

To visualize the proteins a method based on immunohistochemistry was used. A primary antibody recognizes one epitope (monoclonal) or several epitopes (polyclonal) on a protein. In a two-step indirect procedure (figure 6) a visible signal is then created when a labelled secondary antibody reacts with the primary antibody bound to the protein (Boenisch, 1989). In the case of immunofluorescent staining the secondary antibody is conjugated to a fluorescent dye.

Protein

Figure 6. Two step indirect method. The primary antibody (blue) binds to the protein and the secondary antibody (black) conjugated to a fluorescent dye (green) binds to the primary antibody.

The secondary antibody recognizes the origin of the primary antibody. It is consequently possible to label two or more proteins simultaneously if the primary antibodies directed against them are raised in different species.

(16)

Antibodies

To detect DNA-PKCS a mouse monoclonal primary antibody (D4060, Sigma, Saint Lois, Missouri, USA) was used. Mre11 was detected with a rabbit polyclonal primary antibody (PC388, Oncogene, Cambridge, MA, USA). The secondary antibodies were obtained from Molecular Probes (Leiden, The Netherlands). Alexa Fluor® 488 goat anti-mouse (A-11029) and Texas-Red®-X goat anti-rabbit (T-6391) made it possible to detect DNA-PKCS as a green signal and Mre11 as a red signal respectively since Alexa Fluor® 488 emits light with a maximum at 519 nm (green) and Texas-Red®-X emits light with a maximum at 615 nm (red).

Experimental procedure

After repair for various times the cells were briefly washed in serum free medium and fixated in ice cold methanol for 20 minutes at -20˚C in a cuvett. The cells were permeabilized in ice cold acetone for 10 seconds in a cuvett and then washed three times in PBS. Microscopic inspection of the cells was conducted before blocking in 10% FCS-PBS for one hour at room temperature. The cells were washed three times in PBS and then incubated with the two different kinds of primary antibodies mixed in a cocktail. The incubation took place in a humidifying chamber over night at 4˚C.

The antibodies were diluted in 1 % FCS-PBS to a final concentration of 4 µg/ml each.

Following incubation with primary antibodies the cells were washed three times in PBS and incubated with a cocktail of secondary antibodies in a humidifying chamber for approximately one hour at 35-39˚C. The antibodies were diluted in 1 % FCS-PBS to a final concentration of 5 µg/ml each. The cells were washed three times in PBS and then incubated with 0.1 µg/ml DAPI for one minute at room temperature. The treatment with DAPI resulted in a blue staining of the chromatin. Finally the cells were mounted in antifade solution (DAKO, Carpinteria, CA, USA).

Each washing with PBS lasted for a total time of at least 15 minutes in a cuvett. In every incubation step sufficient reagent to cover the cells was used (approximately 200 µl) and reagent was removed between the steps. Drying of cells was avoided throughout the experimental procedure.

As control of background signal the whole immunofluorescent staining was conducted as above, except for the incubation with secondary antibody. The incubation with secondary antibody was replaced with 1 % FCS-PBS.

(17)

The core of the immunofluorescent staining procedure was adapted from Maser et al., 1997.

Microscopy

The microscope used when nothing else is stated was a Leitz DMRXE (Leica, Wetzlar, Germany). The immunofluorescence images were taken with a Hamamatsu ORCA III camera and converted to TIFF format in software Openlab 2.2.0 on a Power Mac G4. Confocal microscopy was done with a Leica TCS-SP and the images were processed using Leica Confocal software. Editing the images was done by using Photoshop 3.0 (Adobe).

(18)

R E S U L T S A N D D I S C U S S I O N

One way to detect intracellular localization of proteins is to analyse occurrence of foci (accumulations). Microscopic inspection of the cells in this study made it possible to detect Mre11 foci with more accuracy than by only analysing the printed pictures.

The results from microscopic inspection are summarized in table 1. All unirradiated controls show 1-2 Mre11 foci in almost half of the cells (data not shown).

Microscopic inspections and printed pictures showed that Mre11, DNA-PKCS, and the chromatin colocalize in the nucleus of the cell (figures 7, 10, 12-14), which is consistent with the fact that Mre11 and DNA-PKCS are nuclear proteins. Furthermore the analysis of the cells showed Mre11 foci after certain repair times but almost never DNA-PKCS foci (figures 7-15). This indicates that the two proteins work in different ways. The lack of Mre11 foci above the level of controls at repair time 0 minutes no matter how the cells were treated (table 1) suggests that Mre11 is spread out in the nucleus and has to be activated and transported before any focus is created. The activation may take some time.

Table 1. Mre11 foci detected by microscopic inspection of a representative part of the whole sample. Cells were exposed to radiation of different kinds of LET.

Repair time

Low LET (10 Gy)

Low LET (10 Gy) + wortmannin

High LET (10 Gy)

High LET (1 Gy)

0 minutes A few foci in some of the cells (fig. 7a).

Granular (fig.

10 a).

1-2 foci in 60% of the cells (fig. 12a).

No sample.

130 minutes

20-35 foci in all of the cells (fig. 7e and 8).

Very granual.

Few foci in all of the cells (fig.

10 e).

1 focus in 20% of the cells (fig. 12e).

5-7 foci in most of the cells (fig. 14a and 15a).

240 minutes

Very granual. A few foci in all of the cells (fig. 7i).

20-35 foci in all of the cells (fig. 9 and 10i).

No sample. No sample.

24 hours 1-2 foci in 30% of the cells, 4 foci in 10% of the cells (fig. 13e).

No sample. 11-25 foci in all of the cells (fig. 11 and 13a).

No sample.

(19)

The Mre11 foci formation is a very interesting aspect of the DSB repair process and an important question is how these foci were affected by LET, dose, and the inhibitor wortmannin.

Cells exposed to 10 Gy low LET (γ-irradiation) showed Mre11 foci formation after 130 minutes (table 1, figure 7e, and figure 8). These foci were not visible after 240 minutes (table 1 and figure 7i) which indicates that the DSB repair is complete by this time or at least that Mre11 has finished its task in the repair process.

Repair- time

(min) Mre11 DNA-PKCS DAPI Merge

Figure 7. Cells exposed to 10 Gy low LET (γ-irradiation) without wortmannin treatment. Images a-d, e-h and i-l (at 40 times magnification) show the same cells respectively.

Cells exposed to 10 Gy low LET in the presence of wortmannin showed a delay of Mre11 foci formation compare to cells exposed to the same dose low LET not treated with wortmannin (table 1 and comparison of figure 7a, e, i and figure 10a, e, i).

0

130

240

(20)

Mre11 foci were detected after 240 minutes of repair in the case of wortmannin treatment (figure 9 and figure 10i). This suggests that the activation of Mre11 is delayed in the presence of wortmannin and, since the inhibitor affects DNA-PKCS, that Mre11 is activated by DNA-PKCS. The delay of Mre11 foci formation could consequently be explained by the action of wortmannin on DNA-PKCS where the inhibition of DNA-PKCS by wortmannin may lead to an obstruction of Mre11 activation.

The Mre11 foci formation after 240 minutes may be due to the loss of activity of wortmannin or alternative pathways for the activation of Mre11. The distribution of DNA-PKCS seem to be unaffected by wortmannin (comparison of figure 7b, f, j and figure 10b, f, j) but its action may still be affected.

Figure 8. Mre11 foci in a cell exposed to 10 Gy low LET without wortmannin treatment. The cell was allowed to recover for 130 minutes. Image was taken with 100 times magnification.

Figure 9. Mre11 foci in a cell exposed to 10 Gy low LET with wortmannin treatment. The cell was allowed to recover for 240 minutes.

Image was taken with 100 times magnification.

(21)

Repair- time

(min) Mre11 DNA-PKCS DAPI Merge

Figure 10. Cells exposed to 10 Gy low LET (γ-irradiation) with wortmannin treatment. Images a-d, e-h and i-l (at 40 times magnification) show the same cells respectively.

When cells were exposed to high LET (accelerated nitrogen ions) there was a different scenario compare to exposure to low LET. At a dose of 10 Gy high LET there were no Mre11 foci at 0 and 130 minutes of repair (table 1, figure 12a and e).

Cells treated with 10 Gy high LET showed very clear foci after 24 hours of repair (figure 11), but cells exposed to low LET did not share this behaviour (table 1 and comparison of figure 13a and e). It should be noted that Mre11 foci formation after long repair time is not due to apoptosis induced DSBs since apoptosis is a rare event in human fibroblasts (Dikomey et al., 1998).

The clear Mre11 foci after 24 hours of repair might instead reflect the slow rejoining of, or inability to rejoin, complex DSBs induced by high LET radiation. 10 Gy of nitrogen Figure 11. Mre11 foci in a cell exposed to

10 Gy high LET and allowed to recover for 24 hours. Image was taken with 100 times magnification.

0

130

240

(22)

ions induces approximately 500 DSBs in a human fibroblast (Höglund et al., 2000).

After 24 hours of repair, 5-10% of these DSBs will still be unrejoined (Stenerlöw et al., 2000), and the number of foci present at this time is within this range.

Repair- time

(min) Mre11 DNA-PKCS DAPI Merge

Figure 12. Cells exposed to 10 Gy high LET (accelerated nitrogen ions). Images a-d and e-h (at 100 times magnification) show the same cells respectively.

Mre11 DNA-PKCS DAPI Merge

Figure 13. Cells exposed to 10 Gy high LET (accelerated nitrogen ions), a-d. Cells exposed to 10 low LET (γ-irradiation), e-h. The cells were allowed to recover for 24 hours. Images were taken with 100 times magnification.

0

130

(23)

Cells exposed to a dose of 1 Gy high LET showed similar Mre11 foci formation after 130 minutes repair as cells exposed to 10 Gy low LET did (table 1 and comparison of figure 7e and figure 14a). When exposed to 1 Gy high LET almost all cells showed 5-7 Mre11 foci (table 1) and this corresponds to the number of nitrogen ions passing the cell nucleus. At a dose of 1 Gy nitrogen ions (LET 125 keV/µm) 7-8 ions pass the average cell nucleus (Höglund and Stenerlöw, in press). In every track of ions 6-8 DSBs are created and subsequently Mre11 is recruited which gives rise to the Mre11 foci formation.

Mre11 DNA-PKCS DAPI Merge

Figure 14. Cells exposed to 1 Gy high LET ( accelerated nitrogen ions) and allowed to recover for 130 minutes. Images were taken with 100 times magnification.

By using a confocal microscope to analyse Mre11 foci in cells exposed to 1 Gy high LET it was possible to detect holes without DNA-PKCS where Mre11 occasionally was located (figure 15). In addition the Mre11 foci and DNA-PKCS holes appeared to elongate through the whole cell nucleus, indicating the passage of a high LET ion.

Mre11 Merge DNA-PKCS

Figure 15. Cell exposed to 1 Gy high LET (perpendicular to the nucleus) and allowed to recover for 130 minutes. The images show the distribution of Mre11 and DNA-PKCS in the nucleus with different projections. The panels are cross-sections of the cell nucleus (the nucleus is thin). The lines intersect in a Mre11 focus (a) and in a DNA-PKCS hole (c). When a and c are merged, the distribution of Mre11 through the cell in the DNA-PKCS hole is obvious (b). A confocal microscope was used, and images were taken with 100 times magnification.

(24)

C O N C L U S I O N S

In conclusion, it seems like Mre11 foci formation is in fact affected by LET and dose.

When cells are exposed to low LET, Mre11 foci are created at an earlier stage in the DSB repair process compare to occurrence of Mre11 foci at exposure to the same dose of high LET. The results further suggest that 1 Gy high LET gives rise to Mre11 foci earlier than exposure to 10 Gy high LET does. A higher dose of low LET and a lower dose of high LET seem to affect the formation of Mre11 foci in a comparable way.

It also seems like the DNA-PK inhibitor wortmannin affects the formation of Mre11 foci. This suggests that DNA-PK activates Mre11. Another correlation between Mre11 and DNA-PKCS is probably the location of the proteins since Mre11 sometimes is located in holes without DNA-PKCS.

Future prospects for this project are to reproduce the results in this report and to investigate Mre11 foci at several repair times after exposure to both high LET and low LET with different doses. The difficulties in analysing the images in an objective way would be somewhat overcome with the access to more samples. Then the conclusions would also be supported in a statistical point of view. It is recommended to use a confocal microscope for analysis in future experiments. A further improvement of the experiments would also be provided with partial irradiation of the cell nucleus, since the distribution of DNA-PKCS and Mre11 would be easier to interpret with more distinct irradiation.

To summarize, with a broader basis of samples (repair times, radiation quality, and dose), partial irradiation, and by using a confocal microscope it will be possible to draw definite conclusions regarding the roles of Mre11 and DNA-PKCS in the DSB repair process.

(25)

R E F E R E N C E S

BOENISCH, T., Staining methods. In Handbook Immunochemical Staining Methods (Naish S.J., Ed.), 13-18. DAKO Corporation, Carpinteria, California (1989).

BOULTON, S., KYLE, S., YALCINTEPE, L. AND DURKACZ, B.W., Wortmannin is a potent inhibitor of DNA double strand break but not single strand break repair in Chinese hamster ovary cells. Carcinogenesis, 17, 2285-2290 (1996).

CARNEY, J.P, MASER, R.S., OLIVARES, H., DAVIS, E.M., LE BEAU, M., YATES, J.R., HAYS, L., MORGAN, W.F. AND PETRINI, J.H.J., The hMre11/hRad50 protein complex and Nijmegen breakage syndrome: linkage of double-strand break repair to the cellular DNA damage response. Cell, 93, 477-486 (1998).

CARY, R.B., PETERSON, S.R., WANG, J., BEAR, D.G., BRADBURY, E.M. AND CHEN, D.J., DNA looping by Ku and the DNA-dependent protein kinase. Proceedings of the National Academy of Sciences of the United States of America, 94, 4267-4272 (1997).

CRITCHLOW, S.E., BOWATER, R.P. AND JACKSON, S.P., Mammalian DNA double- strand break repair protein XRCC4 interacts with DNA ligase IV. Current Biology, 7, 588-598 (1997).

CRITCHLOW, S.E. AND JACKSON, S.P., DNA end-joining: from yeast to man. Trends In Biochemical Sciences, 23, (1998).

DIBIASE, S.J., ZENG, Z-C.,CHEN, R., HYSLOP, T., CURRAN, W.J. AND ILIAKIS, G., DNA- dependent protein kinase stimulates an independently active, nonhomologous, end- joining apparatus. Cancer Research, 60, 1245-1253 (2000).

DIKOMEY, E., DAHM-DAPHI, J., BRAMMER, I., MARTENSEN, R. AND KAINA, B., Correlation between cellular radiosensitivity and non-repaired double-strand breaks studied in nine mammalian cell lines. International Journal of Radiation Biology, 73, 269- 278 (1998).

HABER, J.E., The many interfaces of Mre11. Cell, 95, 583-586 (1998).

HABER, J.E., DNA recombination: the replication connection. Trends In Biochemical Sciences, 24, 271-275 (1999).

HÖGLUND, E., BLOMQUIST, E., CARLSSON, J. AND STENERLÖW, B., DNA damage induced by radiation of different linear energy transfer: initial fragmentation.

International Journal of Radiation Biology, 76, 539-547 (2000).

HÖGLUND, E. AND STENERLÖW, B., Induction and rejoining of DNA damage after exposure to radiation of different linear energy transfer. Possible roles of track structure and chromatin organisation. Radiation Research (in press).

ILIAKIS, G., The role of DNA double strand breaks in ionizing radiation-induced killing of eukaryotic cells. BioEssays, 13, 641-648 (1991).

KARRAN, P., DNA double strand break repair in mammalian cells. Current opinion in Genetics & Development, 10, 144-150 (2000).

(26)

LEUTHER, K.K., HAMMARSTEN,O., KORNBERG, R.D. AND CHU, G., Structure of DNA- dependent protein kinase: implications for its regulation by DNA. The EMBO Journal, 18, 1114-1123 (1999).

MASER, R.S., MONSEN, K.J., NELMS, B.E. AND PETRINI, J.H.J., hMre11 and hRad50 nuclear foci are induced during the normal cellular response to DNA double-strand breaks. Molecular and Cellular Biology, 17, 6087-6096 (1997).

NELMS, B.E., MASER, R.S., MACKAY, J.F., LAGALLY, M.G. AND PETRINI, J.H.J., In situ visualization of DNA double-strand break repair in human fibroblasts. Science, 280, 590-592 (1998).

NIAS, A.H.W., An Introduction to Radiobiology. John Wiley & Sons Ltd, Chichester (1990).

PAULL, T.T. AND GELLERT, M., The 3´ to 5´ exonuclease activity of Mre11 facilitates repair of DNA double-strand breaks. Molecular Cell, 1, 969-979 (1998).

PAULL, T.T., ROGAKOU, E.P., YAMAZAKI, V., KIRCHGESSNER, C.U., GELLERT, M. AND

BONNER, W.M., A critical role for histone H2AX in recruitment of repair factors to nuclear foci after DNA damage. Current Biology, 10, 886-895 (2000).

STENERLÖW, B., BLOMQUIST, E., GRUSELL, E., HARTMAN, T., AND CARLSSON, J., Rejoining of DNA double-strand breaks induced by accelerated nitrogen ions.

International Journal of Radiation Biology, 70, 413-420 (1996).

STENERLÖW, B., HÖGLUND, E., CARLSSON, J. AND BLOMQUIST, E., Rejoining of DNA fragments produced by radiations of different linear energy transfer. International Journal of Radiation Biology, 76, 549-557 (2000).

WARD, J.F., DNA damage produced by ionizing radiation in mammalian cells:

identities, mechanisms of formation, and reparability. Progress in Nucleic Acid Research and Molecular Biology, 35, 95-125 (1988).

(27)

A C K N O W L E D G E M E N T S

I want to express my deepest gratitude to all of you who have supported me during my project work at the division of Biomedical Radiation Sciences. I am especially grateful to my supervisor Ass. Professor Bo Stenerlöw. Your guiding throughout my work has been invaluable, and I really appreciate that you have always had time for my questions.

I am also grateful to Professor Jörgen Carlsson, and to all of you working at BMS for creating such a friendly atmosphere. I have enjoyed the interesting discussions and sharing thoughts with you about the future.

Thank you:

Kenneth Wester, for introducing me to immunological staining procedures.

Irina Radulescu, for assisting me in my practical work.

Stefan Gunnarsson, for contributing with your great knowledge about microscopes and processing of images.

Lars Gedda and Erika Bohl Kullberg, for your help when it comes to get the best images possible!

Finally, I want to thank my wonderful family for always standing by me. Thank you!

This project was supported by grants from the Swedish Cancer Society.

References

Related documents

In a preliminary study, we have compared the P-H2AX foci counting and flow cytometry measurements of P-H2AX in an in vitro-generated mixture of irradiated and non-irradiated

The cumulative effective dose to the foetus from daily internal contamination of 99m Tc (nuclear medicine nurses) and 131 I (cleaning staff) was then estimated in this work by

The aim of this study was to investigate the role of the AKT isoforms AKT1 and AKT2 in cellular response to radiation exposure and their effects on DNA repair proteins (DNA-PKcs

Blir sedan temperaturen vid lagret för hög så smälter smörjmedlet och rinner ut från systemet vilket kommer leda till att lagret skär ihop. Statoil säger att om smörjmedlet

The results showed that Mre11 foci formation was affected by LET and dose, and that the DNA-PK inhibitor wortmannin also has an impact on Mre11 foci formation, suggesting activation

This same technique was used for incorporating both the mutated Rad50 structure and the transition state like structure of Rad50 into the Rad50 wildtype electron density at 2.6

Also the importance of chemically and ionizing radiation induced HLS repair and transformation into DSBs in live cells was assessed in DSB repair inhibited cells. The activation

The design rules for spalling of normal strength concrete, given in Section 6.9.5 may be used for concrete grades C55/67 to C80/95, provided that the maximum content of silica fume