ArticlePDF Available

Live Cell Microscopy of DNA Damage Response in Saccharomyces cerevisiae

Authors:
  • i3S - University of Porto

Abstract and Figures

Fluorescence microscopy of the DNA damage response in living cells stands out from many other DNA repair assays by its ability to monitor the response to individual DNA lesions in single cells. This is particularly true in yeast, where the frequency of spontaneous DNA lesions is relatively low compared to organisms with much larger genomes such as mammalian cells. Single cell analysis of individual DNA lesions allows specific events in the DNA damage response to be correlated with cell morphology, cell cycle phase, and other specific characteristics of a particular cell. Moreover, fluorescence live cell imaging allows for multiple cellular markers to be monitored over several hours. This chapter reviews useful fluorescent markers and genotoxic agents for studying the DNA damage response in living cells and provides protocols for live cell imaging, time-lapse microscopy, and for induction of site-specific DNA lesions.
Content may be subject to copyright.
433
Lotte Bjergbæk (ed.), DNA Repair Protocols, Methods in Molecular Biology, vol. 920,
DOI 10.1007/978-1-61779-998-3_30, © Springer Science+Business Media New York 2012
Chapter 30
Live Cell Microscopy of DNA Damage Response
in Saccharomyces cerevisiae
Sonia Silva , Irene Gallina , Nadine Eckert-Boulet , and Michael Lisby
Abstract
Fluorescence microscopy of the DNA damage response in living cells stands out from many other DNA
repair assays by its ability to monitor the response to individual DNA lesions in single cells. This is particu-
larly true in yeast, where the frequency of spontaneous DNA lesions is relatively low compared to organ-
isms with much larger genomes such as mammalian cells. Single cell analysis of individual DNA lesions
allows speci c events in the DNA damage response to be correlated with cell morphology, cell cycle phase,
and other speci c characteristics of a particular cell. Moreover, uorescence live cell imaging allows for
multiple cellular markers to be monitored over several hours. This chapter reviews useful uorescent mark-
ers and genotoxic agents for studying the DNA damage response in living cells and provides protocols for
live cell imaging, time-lapse microscopy, and for induction of site-speci c DNA lesions.
Key words: Homologous recombination , Checkpoint , Fluorescence microscopy
A broad palette of genetically encoded uorophores have been
developed for uorescence microscopy with the most commonly
used being derivatives of the green and red uorescent proteins
(GFP and RFP) from Aequoria victoria and Discosoma sp., respec-
tively (
1 ) . The available variants of these molecules vary with respect
to excitation and emission spectra, brightness, photostability, pKa,
and oligomeric state in vitro. However, in vivo additional factors
may affect the uorescence capabilities of uorophores. For example,
both GFP and RFP require molecular oxygen for the maturation
of the nascent polypeptide into a chromophore by cyclization and
oxidation of central amino acids before the protein becomes
uorescent (
2, 3 ) . Codon usage is also of great importance for the
proper expression and folding of the uorophore, and notably the
optimal codon usage in Saccharomyces cerevisiae is different from
1. Introduction
434 S. Silva et al.
that of human cells, for which many GFP and RFP variants have
been optimized. Finally, it is advisable to avoid uorophores that
oligomerize because these can compromise biological function of
the fusion protein. We have good experience with three speci c
variants of CFP (clone W7) (
4 ) , YFP (clone 1OC) ( 3 ) and RFP
(clone yEmRFP) (
5 ) . To facilitate PCR-based cloning-free tagging
of proteins at their endogenous chromosomal loci with these
uorophores, we have engineered PCR template vectors that use
the Kluyveromyces lactis URA3 for selection of transformants. The
K. lactis URA3 marker is subsequently popped out to leave a clean
integration of the uorophore encoding open reading frame. The
relevant plasmids (pWJ1162, pWJ1163, pWJ1164, pWJ1165,
pNEB30, and pNEB31) are listed in Table
1 and the tagging pro-
cedure described elsewhere (
13 ) .
The detection of uorescent fusion proteins expressed from
their endogenous promoter depends on multiple factors besides
the expression level including protein turnover, incubation tem-
perature, and localization of the protein. Speci cally, the number
of uorescent molecules available for detection is de ned by the
balance between maturation and protein turnover, which may vary
with experimental conditions. For the CFP, YFP, and RFP variants
used in this study, the maturation half-time was previously esti-
mated to 49, 39, and 15 min, respectively (
14, 15 ) . Moreover,
high temperature negatively affects the folding of some GFP and
RFP variants (
1, 16 ) . In this chapter, we present a selection of
DNA repair and checkpoint markers that are easily detected with
most wide- eld microscopes equipped with a CCD camera
(Subheading
2 , Table 2 and Fig. 1 ). The selected proteins are pri-
marily involved in recombinational DNA repair and checkpoint
activation.
1. SC (synthetic complete) powder: 100 g yeast nitrogen base
without amino acids and without NH
4 SO
4 , 293.11 g NH
4 SO
4 ,
1.2 g adenine sulfate, 1.2 g
L -arginine sulfate, 1.2 g L -histidine-
HCl, 1.8 g
L -isoleucine, 3.6 g L -leucine, 1.8 g L -lysine-HCl,
1.2 g
L -methionine, 3 g L -phenylalanine, 1.2 g L -tryptophan,
1.8 g
L -tyrosine, 1.2 g uracil, 9 g L -valine ( 18 ) . Grind in ball
mill overnight.
2. SC + Ade liquid medium: 7.25 g SC powder, 0.1 g adenine
sulfate, and 20 g glucose in 1,000 mL. Adjust pH to 5.8 with
NaOH/HCl.
3. Galactose: Filter-sterilize 30 % (w/v) stock solution in H
2 O.
2. Materials
2.1. Media and Sugars
43530 Live Cell Microscopy of DNA Damage Response in Saccharomyces cerevisiae
Table 1
Yeast strains and plasmids
Marker Strain/plasmid name Genotype a References
Rad52-G-CFP ML187-2D MAT a TRP1 lys2 D ( 6 )
Rad52-G-CFP pWJ1218 pRS413-based HIS3/
CEN This study
Rad52-G-YFP W5094-1C MAT a trp1-1 LYS2 (
6 )
Rad52-G-YFP pWJ1213 pRS413-based HIS3/
CEN ( 7 )
Rad52-G-YFP pWJ1344 pRS415-based LEU2/
CEN (
7 )
Rad52-G-yEmRFP NEB322 MAT a trp1-1 LYS2 This study
Mec1
1–64 -YFP-AA-Mec1
65–2,369 ML41-4B MAT a trp1-1 LYS2 This study
YFP-GDPGG-Tel1 ML129-6B MAT a trp1-1 LYS2 This study
Ddc2-AAAA-YFP W5088-4A MAT a trp1-1 LYS2 ( 8 )
Ddc2-AAAA-CFP ML311-18C MAT a TRP1 lys2 D This study
Mre11-LLAKKRKG-YFP ML104-6B MAT a trp1-1 LYS2 ( 8 )
Rfa1-AAAAAAAAG-YFP ML147-2B MAT a trp1-1 LYS2 ( 8 )
Rfa1-AAAAAAAAG-CFP ML67-4B MAT a TRP1 lys2 D This study
YFP-AAAAAAAA-Sml1 W4622-14D MAT a trp1-1 LYS2
bar1::LEU2 ( 9 )
Ddc1-AAAA-YFP W5059-8A MAT a trp1-1 LYS2 This study
YFP-GGPGG-Rad9 SMG242 MAT a trp1-1 LYS2 (
10 )
Mec1
1-64 -YFP-AA-Mec1
65-2,369 ML292-6C MAT a trp1-1 LYS2 This study
Ddc2-AAAA-CFP
YFP-AAAAAAAA-Sml1 IG101-12D MAT a trp1-1 LYS2 This study
Rad52-G-yEmRFP
Mre11-LLAKKRKG-YFP SS54-9B MAT a trp1-1 LYS2
RAD52-RFP This study
Rfa1-AAAAAAAAG-CFP
CFP-3 ¢ -K.l.URA3 pWJ1162 pRS423-based HIS3/2 m (
11 )
CFP-5 ¢ -K.l.URA3 pWJ1163 pRS423-based HIS3/2 m (
11 )
YFP-3 ¢ -K.l.URA3 pWJ1164 pRS423-based HIS3/2 m (
11 )
YFP-5 ¢ -K.l.URA3 pWJ1165 pRS423-based HIS3/2 m (
11 )
yEmRFP-3 ¢ -K.l.URA3 pNEB31 pRS423-based HIS3/2 m This study
yEmRFP-5 ¢ -K.l.URA3 pNEB30 pRS423-based HIS3/2 m This study
a All strains used in this study are ADE2 RAD5 derivatives of W303-1A ( BAR1 ade2-1 can1-100 ura3-1 his3-11,15 leu2-
3,112 trp1-1 LYS2 rad5-535 ) ( 12 )
436 S. Silva et al.
Table 2
DNA damage response markers, exposure time, and biological function
Protein Construct a Biological function Localization
Exposure
time
b References
Rad52 Rad52-G-CFP, Rad52-G-
YFP, Rad52-G-yEmRFP Early marker of HR Diffuse nuclear, partially excluded from the
nucleolus. Relocalizes to foci during HR 1 s ( 5, 6 )
Mec1 Mec1
1–64 -YFP-AA-
Mec1
65–2,369 Single-stranded DNA
damage checkpoint Localizes from diffuse nuclear to foci in
response to DNA damage 3 s (
8 )
Tel1 YFP-GDPGG-Tel1 Double-strand break
checkpoint Localizes from diffuse nuclear to foci in
response to DSBs 7 s (
17 )
Ddc2 Ddc2-AAAA-YFP Single-stranded DNA
damage checkpoint Localizes from diffuse nuclear to foci in
response to DNA damage 3 s (
17 )
Ddc2 Ddc2-AAAA-CFP Single-stranded DNA
damage checkpoint Localizes from diffuse nuclear to foci in
response to DNA damage 1 s (
17 )
Mre11 Mre11-LLAKKRKG-YFP Double-strand break
recognition and processing Localizes from diffuse nuclear to foci in
response to DSBs 3 s (
17 )
Rfa1 Rfa1-AAAAAAAAG-CFP Single-stranded DNA
recognition Multiple faint replication foci during S phase.
Localizes to bright foci in response to DSBs 0.5 s (
17 )
Sml1 YFP-AAAAAAAA-Sml1 Downstream target of
Rad53-dependent DNA
damage checkpoint
Cytoplasmic degraded upon checkpoint
activation and during S phase 3 s (
8, 9 )
Ddc1 Ddc1-AAAA-YFP Single-stranded DNA
damage checkpoint Localizes from diffuse nuclear to faint foci in
response to DNA damage 3 s (
17 )
Rad9 YFP-GGPGG-Rad9 DNA damage checkpoint Localizes from diffuse nuclear to transient
foci in response to DNA damage 3 s (
10 )
a For microscopy of a single protein, YFP is the preferred uorophore for tagging, because it gives the best signal-to-noise ratio. For imaging three proteins in the same cell, it is advis-
able to tag the protein of the lowest abundance with YFP to maximize sensitivity, the most abundant protein with CFP to reduce phototoxicity, and the third protein with RFP
b The optimal exposure time depends on the uorophore, the abundance of the protein, the sensitivity of the camera and the intensity of the excitation light, which should be
adjusted so that no signi cant photobleaching occurs during acquisition
43730 Live Cell Microscopy of DNA Damage Response in Saccharomyces cerevisiae
1. Melt and mix 1 volume of petroleum jelly (vaseline), 1 volume
of beeswax, and 1 volume of lanolin at 80 °C.
2. For sealing the edges of the microscope cover glass for time-
lapse microscopy, the wax mixture can be kept at 65 °C. The
molten wax is applied along the edges of the cover glass with a
at metal spatula pre-heated over a Bunsen burner or similar.
The following chemicals were used: zeocin, camptothecin (CPT),
methyl methanesulfonate (MMS), raf nose, galactose, petroleum
jelly, beeswax, lanolin, 4-nitroquinoline-1-oxide (4-NQO), and
hydroxyurea (HU).
The essential elements of the microscope hardware are a high sen-
sitivity cooled CCD or EM-CCD camera and a high magni cation
objective (100×) with high numerical aperture ( ³ 1.4).
2.2. Wax for Sealing
Microscope Slides
2.3. Reagents
and Chemicals
2.4. Microscope
Hardware
Fig. 1. Representative images of selected DNA repair and checkpoint proteins. Strains
were imaged as described in Subheading
3 after treatment with 100 m g/mL zeocin for
1 h. Strains are ( a ) ML292-6C, ( b ) IG101-12D, ( c ) ML129-6B, ( d ) W5059-8A, ( e ) SMG242,
and ( f ) SS54-9B. Scale bar, 3 µm.
438 S. Silva et al.
For yeast live-cell imaging, the best result is obtained by culturing
and mounting cells in lter-sterilized minimal (SC) medium. Live
cell imaging is preferred over xed cells, because xation may gen-
erate artifacts or otherwise decrease the quality of the obtained
data. A protocol for imaging live yeast cells is described here:
1. Inoculate from a single colony 3 mL of SC medium containing
100 m g/mL adenine (see Note 1).
2. Grow shaking overnight at 25 °C (see Note 2).
3. Dilute culture to OD
600 = 0.2 and grow for at least one cell
cycle (2.5 h for wild type) prior to microscopy.
4. Expose the culture to DNA damage using one of the agents in
Table
3 .
5. Harvest 1.5 mL of cells at OD
600 = 0.4–0.6 by centrifugation at
1,500 × g (see Note 3).
6. Resuspend the pellet of cells in 50 m L of medium by
vortexing.
7. Cells are mounted on standard glass slides and covered by
cover glass appropriate to the optics of the microscope (see
Note 4). For most strains, the cells are immobilized on the
slide simply by adjusting the volume applied (usually 2–3 m L),
so that the cells settle in a monolayer with the cells touching
both the slide and the cover glass.
8. Capture 11 optical sections separated by 0.4 m m around the
focal plane of the cell (see Note 5).
Imaging of cells over time requires that phototoxicity is minimized
and that favorable growth conditions can be maintained (see Note
6). Phototoxicity can be reduced by decreasing the uorescence
exposure time or intensity of the excitation light, and by reducing
the number of optical sections acquired. The easiest way to main-
tain favorable growth conditions is to reduce the cell density on
the slide, so that only a single cell is present in the eld of view,
thereby prolonging the time that the cell can grow before nutrients
are locally exhausted. For this reason it is not recommended to
concentrate the cells by centrifugation before mounting. For long-
term imaging (>30 min), the edges of the cover glass are sealed
with wax to prevent evaporation (see Subheading
2.2 and Note 7).
The protocol is:
1. Turn on the microscope 1–2 h before the time-lapse is launched
(see Note 8).
2. Cells are cultured and mounted essentially as described above
(see Subheading
3.2 ).
3. Methods
3.1. Basic Imaging
Protocol
for Fluorescence
Microscopy
3.2. Time-Lapse
Microscopy
43930 Live Cell Microscopy of DNA Damage Response in Saccharomyces cerevisiae
3. The appropriate acquisition parameters for time-lapse
microscopy should be determined empirically. With our
microscopy setup, we are able to image Rad52-YFP with
minimal phototoxicity for 20 time points over 4 h by reduc-
ing the number of optical sections from 11 to 9 (0.5 m m
between sections), by reducing the intensity of the excita-
tion light by inserting a neutral density lter in the
uorescence path that allows only 10 % transmission, and by
reducing the exposure time from 1 to 0.5 s.
Table 3
Compounds for DNA damage induction
Genotoxic
agent
a Dosage
Time of
treatment Targets/lesions
Primary repair
pathway(s) References
HU 100–200 mM 30 min Inhibits ribonucle-
otide reductase
leading to depletion
of dNTP pools and
replication fork
stalling
Replication restart
by homologous
recombination
(
19, 20 )
MMS 0.03 % 1 h Methylates DNA on
N
7 -deoxyguanine
and N
3 -
deoxyadenine,
which leads to
replication fork
stalling and collapse
Replication restart
by homologous
recombination,
DSB repair
(
21, 22 )
IR 1 DSB per
haploid
genome per
2 krad (20 Gy)
Seconds–
minutes In addition to DSBs,
approximately 17
SSBs are generated
per DSB
Homologous
recombination (
23 )
4-NQO 10 ng/mL 1 h Guanine adducts at
positions C
8 and N
2 Nucleotide
excision repair,
translesion
synthesis, HR
(
24, 25 )
CPT 5 m g/mL 1 h Stabilizes covalent
topoisomerase
I-DNA intermedi-
ates, which are
converted to DSBs
during DNA
replication
Excision repair and
homologous
recombination
(
26, 27 )
Zeocin 100 m g/mL 1 h DSBs Homologous
recombination (
28 )
a HU hydroxyurea, MMS methyl methanesulfonate, IR ionizing radiation, 4-NQO 4-nitroquinoline-1-oxide, CPT
camptothecin, SSB single-strand break, DSB double-strand break, HR homologous recombination
440 S. Silva et al.
The advantage of site-speci c, inducible DNA lesions is that the
location of the lesion can be marked uorescently and therefore
followed during repair. Fluorescence marking of the DNA lesion is
achieved by inserting a tandem array of DNA binding sites for a
uorescently tagged protein, e.g., the Lac or Tet repressor, adja-
cent to the DNA lesion as described (
13 ) . Different types of site-
speci c DNA lesions can be introduced. For example, the HO and
I-SceI meganucleases are used to induce double-strand breaks
(DSBs) (
21 ) ; a mutant of the Flp recombinase (Flp-H305L) irre-
versibly cleaves one DNA strand forming a covalent Flp-DNA
intermediate (
29 ) , which causes replication fork stalling and col-
lapse; and a mutant of the I-AniI homing endonuclease (I-AniI-
K227M) exhibits site-speci c nicking (
30 ) . Strains in which a single
site-speci c endonuclease restriction site in the genome has been
marked uorescently, such as the HO or the I-SceI sites, are trans-
formed with a plasmid expressing the appropriate endonuclease
from an inducible promoter, e.g., pJH132 (pGAL-HO) or
pWJ1108 (pGAL-I-SceI), respectively (
8, 31 ) .
1. Transform the appropriate plasmid (expressing HO or I-SceI)
into the assay strain by the LiAc method (
32 ) . Select trans-
formants on the appropriate glucose-containing minimal
medium.
2. Inoculate fresh transformants in 2 mL of the appropriate mini-
mal, raf nose-based medium (2 %, w/v, raf nose, autoclaved)
(see Note 9).
3. Grow shaking at 25 °C for 24 h.
4. Dilute culture to OD
600 = 0.2 and grow for one cell cycle (3.5 h
for wild type) prior to endonuclease induction.
5. Add galactose (sterile- ltered, stock solution 30 %, w/v) to a
nal concentration of 2–3 % (w/v). This induces expression of
the endonuclease.
6. Grow shaking at 25 °C for another 90 min prior to microscopy.
1. The supplementation of SC medium with 100 m g/mL adenine
is necessary only for ade2 mutant strains, which will otherwise
accumulate a red pigment that is strongly auto uorescent.
However, this medium can also be used for ADE2 wild-type
cells.
2. It is important that the culture is well aerated, because matura-
tion of GFP and RFP requires molecular oxygen (
2, 3 ) .
3.3. Site-Speci fi c
DNA Lesions
4. Notes
44130 Live Cell Microscopy of DNA Damage Response in Saccharomyces cerevisiae
3. At this point it is not necessary to use autoclaved microcentrifuge
tubes. In fact, yeast cells tend to adhere to the side of some
microcentrifuge tubes, if they have been autoclaved, whereas
cells will pellet to the bottom of untreated tubes.
4. A high cell density in the eld of view is desired to maximize
data acquisition. However, at high cell density and growth rate
(glucose), the medium quickly becomes saturated with carbon
dioxide, which precipitates as gas bubbles that may displace
cells. Therefore, the optimal cell density must often be deter-
mined empirically.
5. The yeast nucleus has a diameter of approximately 2 m m and
the cell 5 m m. The number of optical sections required to sam-
ple the entire volume of the cell is determined by the Nyquist
theorem. Ideally, the sampling distance should be half the
depth of focus, but in our experience a sampling distance of
0.3–0.4 m m is suf cient for most applications. However, under-
sampling may cause some structures to be overlooked.
6. Phototoxicity is of particular importance when studying DNA
damage response, because photolesions can be introduced
into the DNA especially when imaging CFP, which has the
excitation wavelength with the highest energy compared to
YFP and RFP.
7. Nail polish or other solutions containing organic solvents
should be avoided when sealing slides for live cell imaging,
because the solvents may harm the cells.
8. Most microscopes will heat up and expand during use, which
will cause the focal plane to change. The temperature of the
microscope will usually reach equilibrium within 1–2 h of use.
9. It is important to use fresh transformants in order to avoid
selecting cells in which mutations have accumulated in the
endonuclease recognition site. Such mutations tend to
appear in old transformants due to leakage of the GAL1-10
promoter.
Acknowledgment
We thank Dr. Neta Dean, Stony Brook University, for sharing the
yEmRFP construct. This work was supported by Fundação para a
Ciência e a Tecnologia (SS), The Danish Agency for Science,
Technology and Innovation (ML, NEB), the Villum Kann Rasmussen
Foundation (ML), and the European Research Council (ML).
442 S. Silva et al.
References
1. Shaner NC, Steinbach PA, Tsien RY (2005) A
guide to choosing uorescent proteins. Nat
Methods 2:905–909
2. Remington SJ (2006) Fluorescent proteins:
maturation, photochemistry and photophysics.
Curr Opin Struct Biol 16:714–721
3. Ormo M, Cubitt AB, Kallio K, Gross LA,
Tsien RY, Remington SJ (1996) Crystal
structure of the Aequorea victoria green
uorescent protein. Science 273:1392–1395
4. Heim R, Tsien RY (1996) Engineering green
uorescent protein for improved brightness,
longer wavelengths and uorescence resonance
energy transfer. Curr Biol 6:178–182
5. Keppler-Ross S, Noffz C, Dean N (2008) A
new purple uorescent color marker for genetic
studies in Saccharomyces cerevisiae and Candida
albicans . Genetics 179:705–710
6. Lisby M, Rothstein R, Mortensen UH (2001)
Rad52 forms DNA repair and recombination
centers during S phase. Proc Natl Acad Sci
U S A 98:8276–8282
7. Alvaro D, Sunjevaric I, Reid RJ, Lisby M,
Stillman DJ, Rothstein R (2006) Systematic
hybrid LOH: a new method to reduce false
positives and negatives during screening of yeast
gene deletion libraries. Yeast 23:1097–1106
8. Torres-Rosell J, Sunjevaric I, De Piccoli G,
Sacher M, Eckert-Boulet N, Reid R, Jentsch S,
Rothstein R, Aragon L, Lisby M (2007) The
Smc5-Smc6 complex and SUMO modi cation
of Rad52 regulates recombinational repair at the
ribosomal gene locus. Nat Cell Biol 9:923–931
9. Andreson BL, Gupta A, Georgieva BP,
Rothstein R (2010) The ribonucleotide
reductase inhibitor, Sml1, is sequentially phos-
phorylated, ubiquitylated and degraded in
response to DNA damage. Nucleic Acids Res
38:6490–6501
10. Germann SM, Oestergaard VH, Haas C, Salis
P, Motegi A, Lisby M (2011) Dpb11/TopBP1
plays distinct roles in DNA replication, check-
point response and homologous recombina-
tion. DNA Repair (Amst) 10:210–224
11.
Reid R, Lisby M, Rothstein R (2002) Cloning-
free genome alterations in Saccharomyce cerevi-
siae using adaptamer-mediated PCR. Methods
Enzymol 350:258–277
12. Zhao X, Muller EG, Rothstein R (1998) A sup-
pressor of two essential checkpoint genes
identi es a novel protein that negatively affects
dNTP pools. Mol Cell 2:329–340
13. Eckert-Boulet N, Rothstein R, Lisby M (2011)
Cell biology of homologous recombination in
yeast. Methods Mol Biol 745:523–536
14. Gordon A, Colman-Lerner A, Chin TE,
Benjamin KR, Yu RC, Brent R (2007) Single-
cell quanti cation of molecules and rates using
open-source microscope-based cytometry. Nat
Methods 4:175–181
15. Shaner NC, Campbell RE, Steinbach PA,
Giepmans BN, Palmer AE, Tsien RY (2004)
Improved monomeric red, orange and yellow
uorescent proteins derived from Discosoma sp.
red uorescent protein. Nat Biotechnol 22:
1567–1572
16. Lim CR, Kimata Y, Oka M, Nomaguchi K,
Kohno K (1995) Thermosensitivity of green
uorescent protein uorescence utilized to
reveal novel nuclear-like compartments in a
mutant nucleoporin NSP1. J Biochem (Tokyo)
118:13–17
17. Lisby M, Barlow JH, Burgess RC, Rothstein R
(2004) Choreography of the DNA damage
response; spatiotemporal relationships among
checkpoint and repair proteins. Cell 118:
699–713
18. Sherman F, Fink GR, Hicks JB (1986) Methods
in yeast genetics. Cold Spring Harbor
Laboratory, Cold Spring Harbor, NY
19. Reichard P (1988) Interactions between deoxy-
ribonucleotide and DNA synthesis. Annu Rev
Biochem 57:349–374
20. Lopes M, Cotta-Ramusino C, Pellicioli A,
Liberi G, Plevani P, Muzi-Falconi M, Newlon
CS, Foiani M (2001) The DNA replication
checkpoint response stabilizes stalled replica-
tion forks. Nature 412:557–561
21. Lisby M, Mortensen UH, Rothstein R (2003)
Colocalization of multiple DNA double-strand
breaks at a single Rad52 repair centre. Nat Cell
Biol 5:572–577
22.
Beranek DT, Weis CC, Swenson DH (1980) A
comprehensive quantitative analysis of methy-
lated and ethylated DNA using high pressure
liquid chromatography. Carcinogenesis 1:
595–606
23. Friedland W, Jacob P, Paretzke HG, Merzagora
M, Ottolenghi A (1999) Simulation of DNA
fragment distributions after irradiation with
photons. Radiat Environ Biophys 38:39–47
24. Fronza G, Campomenosi P, Iannone R,
Abbondandolo A (1992) The 4-nitroquinoline
1-oxide mutational spectrum in single stranded
DNA is characterized by guanine to pyrimidine
transversions. Nucleic Acids Res 20:1283–1287
25. Wiltrout ME, Walker GC (2011) The DNA
polymerase activity of Saccharomyces cerevisiae
Rev1 is biologically signi cant. Genetics 187:
21–35
44330 Live Cell Microscopy of DNA Damage Response in Saccharomyces cerevisiae
26. Deng C, Brown JA, You D, Brown JM (2005)
Multiple endonucleases function to repair
covalent topoisomerase I complexes in
Saccharomyces cerevisiae . Genetics
170:591–600
27. Eng WK, Faucette L, Johnson RK, Sternglanz
R (1988) Evidence that DNA topoisomerase I
is necessary for the cytotoxic effects of camp-
tothecin. Mol Pharmacol 34:755–760
28. Moore CW, McKoy J, Dardalhon M,
Davermann D, Martinez M, Averbeck D
(2000) DNA damage-inducible and RAD52 -
independent repair of DNA double-strand
breaks in Saccharomyces cerevisiae . Genetics
154:1085–1099
29. Nielsen I, Bentsen IB, Lisby M, Hansen S,
Mundbjerg K, Andersen AH, Bjergbaek L
(2009) A Flp-nick system to study repair of a
single protein-bound nick in vivo. Nat Methods
6:753–757
30. McConnell Smith A, Takeuchi R, Pellenz S,
Davis L, Maizels N, Monnat RJ Jr, Stoddard BL
(2009) Generation of a nicking enzyme that
stimulates site-speci c gene conversion from the
I-AniI LAGLIDADG homing endonuclease.
Proc Natl Acad Sci U S A 106:5099–5104
31. Jensen RE, Herskowitz I (1984) Directionality
and regulation of cassette substitution in yeast.
Cold Spring Harbor Symp Quant Biol
49:97–104
32. Gietz D, St Jean A, Woods RA, Schiestl RH
(1992) Improved method for high ef ciency
transformation of intact yeast cells. Nucleic
Acids Res 20:1425
... Seminal time-lapse imaging studies used protein fusions to elucidate the spatio-temporal relationships between the budding yeast DNA damage checkpoint and repair proteins [1][2][3]. The timing and localization of these proteins remain useful reporters in live cell studies of the DNA repair process [4,5]. Alternatively, fluorescent proteins can be used as transcriptional reporters by fusing their coding sequence to a promoter of interest [6,7]. ...
... 3. Degas PDMS mixture for % 30 min in a desiccator, until solution is free of bubbles (see Note 7). 4. Place wafer inside the Petri dish on a scale and pour 25 g of degassed PDMS onto the wafer. ...
... Exposure to heat and sunlight will be detrimental for adhesion of the SU-8 on the wafer. 4. The microfluidic device can be re-used as often as % 30 times. ...
Chapter
Time-lapse fluorescence imaging of yeast cells allows the study of multiple fluorescent targets in single cells, but is often hampered by the tedious cultivation using agar pads or glass bottom wells. Here, we describe the fabrication and operation of a microfluidic device for long-term imaging of yeast cells under constant or changing media conditions. The device allows acquisition of high quality images as cells are fixed in a two-dimensional imaging plane. Four yeast strains can be analyzed simultaneously over several days while up to four different media can be flushed through the chip. The microfluidic device does not rely on specialized equipment for its operation. To illustrate the use of the chip in DNA damage research, we show how common readouts for DNA damage or genomic instability behave upon induction with genotoxic chemicals (MMS, HU) or induction of a single double-strand break using induced CRISPR-Cas9 expression.
... The slide was then covered with a cover glass, and slight pressure was applied. All procedures were carried out at 27 • C and according to the protocol described by Silva and co-workers [52]. For some of the experiments, HU was added to the solid and liquid media. ...
Article
Full-text available
Cells have evolved elaborate mechanisms to regulate DNA replication machinery and cell cycles in response to DNA damage and replication stress in order to prevent genomic instability and cancer. The E3 ubiquitin ligase SCFDia2 in S. cerevisiae is involved in the DNA replication and DNA damage stress response, but its effect on cell growth is still unclear. Here, we demonstrate that the absence of Dia2 prolongs the cell cycle by extending both S- and G2/M-phases while, at the same time, activating the S-phase checkpoint. In these conditions, Ctf4—an essential DNA replication protein and substrate of Dia2—prolongs its binding to the chromatin during the extended S- and G2/M-phases. Notably, the prolonged cell cycle when Dia2 is absent is accompanied by a marked increase in cell size. We found that while both DNA replication inhibition and an absence of Dia2 exerts effects on cell cycle duration and cell size, Dia2 deficiency leads to a much more profound increase in cell size and a substantially lesser effect on cell cycle duration compared to DNA replication inhibition. Our results suggest that the increased cell size in dia2∆ involves a complex mechanism in which the prolonged cell cycle is one of the driving forces.
... The two homology arms were fused by PCR to sequences containing yEmRFP joined to either the 5'-or 3'-end of K.l. URA3 that were PCR amplified from pNEB30 or pNEB31 (Silva et al, 2012) using primers Cherry.Fw and 3'-int or 5'-int and yEmRFP-R. The two fusion products were co-transformed into IG147 and selected on SC-Ura. ...
Preprint
DNA replication stress is a source of genome instability and a replication checkpoint has evolved to enable fork stabilisation and completion of replication during stress. Mediator of the replication checkpoint 1 (Mrc1) is the primary mediator of this response in Saccharomyces cerevisiae. Mrc1 is partially sequestered in the intranuclear quality control compartment (INQ) upon methyl methanesulfonate (MMS)-induced replication stress. Here we show that Mrc1 re-localizes from the replication fork to INQ during replication stress. Sequestration of Mrc1 in INQ is facilitated by the Btn2 chaperone and the Cdc48 segregase is required to release Mrc1 from INQ during recovery from replication stress. Consistently, we show that Cdc48 colocalizes with Mrc1 in INQ and we find that Mrc1 is recognized by the Cdc48 cofactors Ufd1 and Otu1, which contribute to clearance of Mrc1 from INQ. Our findings suggest that INQ localization of Mrc1 and Cdc48 function to facilitate replication stress recovery by transiently sequestering the replication checkpoint mediator Mrc1 and explains our observation that Btn2 and Cdc48 are required for efficient replication restart following MMS-induced replication stress.
... For live cell microscopy of Rfa1 and Ddc1 recruitment to an HO-induced DSB, cells were grown shaking in liquid SC+Ade medium (synthetic complete medium supplemented with 100 µg/ml adenine) with 2% raffinose at 25°C to OD600 = 0.2-0.3 and arrested either in G1 phase with 10 µg/ml α-factor or in M phase with 15 µg/ml nocodazole for 2 h before addition of galactose to a final concentration of 2%. Cells were processed for fluorescence microscopy at the indicated times after addition of galactose as established 74 . Fluorophores were visualized on a Deltavision Elite microscope (Applied Precision, Inc) equipped with a 100× objective lens (Olympus U-PLAN S-APO, NA 1.4), a cooled Evolve 512 EMCCD camera (Photometrics, Japan) and an Insight solid-state illumination source (Applied Precision, Inc.). ...
Article
Full-text available
The DNA damage checkpoint senses the presence of DNA lesions and controls the cellular response thereto. A crucial DNA damage signal is single-stranded DNA (ssDNA), which is frequently found at sites of DNA damage and recruits the sensor checkpoint kinase Mec1-Ddc2. However, how this signal – and therefore the cell's DNA damage load – is quantified, is poorly understood. Here, we use genetic manipulation of DNA end resection to induce quantitatively different ssDNA signals at a site-specific double strand break in budding yeast and identify two distinct signalling circuits within the checkpoint. The local checkpoint signalling circuit leading to γH2A phosphorylation is unresponsive to increased amounts of ssDNA, while the global checkpoint signalling circuit, which triggers Rad53 activation, integrates the ssDNA signal quantitatively. The global checkpoint signal critically depends on the 9-1-1 and its downstream acting signalling axis, suggesting that ssDNA quantification depends on at least two sensor complexes.
... it easier to identify cells with two foci that are separated by more than 1 µm, which is the minimum distance required to photobleach one focus without collateral bleaching of the adjacent focus with most microscope configurations. Under standard time-lapse imaging conditions [290], twothree DSBs per diploid cell can be induced by treatment with 200 µg/ml Zeocin for 2 h or 30 Gy of ionizing radiation. After this treatment, 10-20% of the cells will have two foci of for example the Rad52 recombination mediator or the single-stranded DNA binding protein RPA (Replication protein A) [291], while most of the remaining cells have one focus. ...
Article
Full-text available
Understanding the plasticity of genomes has been greatly aided by assays for recombination, repair and mutagenesis. These assays have been developed in microbial systems that provide the advantages of genetic and molecular reporters that can readily be manipulated. Cellular assays comprise genetic, molecular, and cytological reporters. The assays are powerful tools but each comes with its particular advantages and limitations. Here the most commonly used assays are reviewed, discussed, and presented as the guidelines for future studies.
... Strains used for microscopy are ADE2 LYS2 trp1-1 derivatives of W1588-4C [68], a RAD5 derivate of W303-1A (MATa ade2-1 can1-100 ura3-1 his3-11,15 leu2-3,112 trp1-1 rad5-535) [69]. CFPtagged Mph1 was generated for expression from its native chromosomal locus with a 4-alanine linker as described [70,71] using primers listed in S1 Table and verified by sequencing. The HPR1 gene was deleted with KANMX6 in this strain to construct YBG722. ...
Article
Full-text available
Author summary DNA damage can either occur exogenously through DNA damaging agents such as UV light and exposure to chemotherapeutics, or endogenously via metabolic, cellular processes. The RNA product of transcription, for example, can engage in the formation of RNA-DNA hybrids. Such RNA-DNA hybrids can impede replication fork progression and cause genomic instability, a hallmark of cancer. The misregulation of RNA-DNA hybrids has also been implicated in several neurological disorders. Recently, it has become evident that RNA-DNA hybrids may also have beneficial roles and therefore, these structures have to be tightly controlled. We found that Mph1 (mutator phenotype 1), the budding yeast homolog of Fanconi Anemia protein M, counteracts the accumulation of RNA-DNA hybrids. The inactivation of MPH1 results in a severe growth defect when combined with mutations in the well-characterized RNase H enzymes, that degrade the RNA moiety of an RNA-DNA hybrid. Based on the data presented here, we propose a model, where Mph1 itself has to be kept in check by the SMC (structural maintenance of chromosome) 5/6 complex at replication forks stalled by RNA-DNA hybrids. Mph1 acts as a double-edged sword, as both its deletion and the inability to control its helicase activity cause DNA damage and growth arrest when RNA-DNA hybrids accumulate.
... Most strains were grown overnight in air orbital incubators at 25º in YPD media before every experiment. Cell cycle time course experiments and fluorescence microscopy were performed as described before (Silva et al. 2012;Quevedo et al. 2012;. Briefly, asynchronous cultures of MATa haploids were adjusted to OD 600 = 0.3 and then synchronized in G1 at 25º for 3 h by adding 50 ng/ml (bar1Δ strains) or 5 μg/ml (BAR1 strains) of α-factor (T6901, Sigma-Aldrich). ...
Article
Full-text available
Topoisomerase II (Top2) is the essential protein that resolves DNA catenations. When Top2 is inactivated, mitotic catastrophe results from massive entanglement of chromosomes. Top2 is also the target of many first-line anticancer drugs, the so-called Top2 poisons. Often, tumors become resistant to these drugs by acquiring hypomorphic mutations in the genes encoding Top2. Here, we have compared the cell cycle and nuclear segregation of two coisogenic Saccharomyces cerevisiae strains carrying top2 thermosensitive alleles that differ in their resistance to Top2 poisons: the broadly-used poison-sensitive top2-4 and the poison-resistant top2-5 Furthermore, we have performed genome-scale Synthetic Genetic Array (SGA) analyses for both alleles under permissive conditions, chronic sublethal Top2 downregulation and acute, yet transient, Top2 inactivation. We find that slowing down mitotic progression, especially at the time of execution of the Mitotic Exit Network (MEN), protects against Top2 deficiency. In all conditions, genetic protection was stronger in top2-5, and this correlated with cell biology experiments in this mutant whereby we observed destabilization of both chromatin and ultrafine anaphase bridges by execution of MEN and cytokinesis. Interestingly, whereas transient inactivation of the critical MEN driver Cdc15 partly suppressed top2-5 lethality, this was not the case when earlier steps within anaphase were disrupted; i.e., top2-5 cdc14-1 We discuss the basis of this difference and suggest that accelerated progression through mitosis may be a therapeutic strategy to hypersensitize cancer cells carrying hypomorphic mutations in TOP2.
Article
Full-text available
Dna2 is an essential nuclease-helicase that acts in several distinct DNA metabolic pathways including DNA replication and recombination. To balance these functions and prevent unscheduled DNA degradation, Dna2 activities must be regulated. Here we show that Saccharomyces cerevisiae Dna2 function is controlled by sumoylation. We map the sumoylation sites to the N-terminal regulatory domain of Dna2 and show that in vitro sumoylation of recombinant Dna2 impairs its nuclease but not helicase activity. In cells, the total levels of the non-sumoylatable Dna2 variant are elevated. However, non-sumoylatable Dna2 shows impaired nuclear localization and reduced recruitment to foci upon DNA damage. Non-sumoylatable Dna2 reduces the rate of DNA end resection, as well as impedes cell growth and cell cycle progression through S phase. Taken together, these findings show that in addition to Dna2 phosphorylation described previously, Dna2 sumoylation is required for the homeostasis of the Dna2 protein function to promote genome stability. Ranhja et al. find that budding yeast Dna2 is modified by sumoylation, and that sumoylation reduces the nuclease, but not helicase activity of Dna2 in vitro. In cells, expression of a Dna2 version that cannot be sumoylated leads to impaired DNA end resection and cell division.
Article
Full-text available
Dna2 is a nuclease and helicase that functions redundantly with other proteins in Okazaki fragment processing, double strand break (DSB) resection and checkpoint kinase activation. Dna2 is an essential enzyme, required for yeast and mammalian cell viability. Here we report that numerous mutations affecting the DNA damage checkpoint suppressdna2Δlethality inSaccharomyces cerevisiaedna2Δcells are also suppressed by deletion of helicases,PIF1andMPH1, and by deletion ofPOL32, a subunit of DNA polymerase δ. Alldna2Δcells are temperature sensitive, have telomere length defects, and low levels of telomeric 3' single stranded DNA (ssDNA). Interestingly, Rfa1, a subunit of the major ssDNA binding protein RPA, and the telomere specific ssDNA binding protein Cdc13, often co-localize indna2Δcells. This suggests that telomeric defects often occur indna2Δ cells. There are several plausible explanations for why the most critical function of Dna2 is at telomeres. Telomeres modulate the DNA damage response (DDR) at chromosome ends, inhibiting resection, ligation and cell cycle arrest. We suggest that Dna2 nuclease activity contributes to modulating the DNA damage response at telomeres by removing telomeric C-rich ssDNA and thus preventing checkpoint activation.
Chapter
DNA ultrafine bridges (UFBs) are a type of chromatin-free DNA bridges that connect sister chromatids in anaphase and pose a threat to genome stability. However, little is known about the origin of these structures, and how they are sensed and resolved by the cell. In this chapter, we review tools and methods for studying UFBs by fluorescence microscopy including chemical and genetic approaches to induce UFBs in the model organism Saccharomyces cerevisiae.
Article
Full-text available
Homologous recombination (HR) is crucial for maintaining genome integrity by repairing DNA double-strand breaks (DSBs) and rescuing collapsed replication forks. In contrast, uncontrolled HR can lead to chromosome translocations, loss of heterozygosity, and deletion of repetitive sequences. Controlled HR is particularly important for the preservation of repetitive sequences of the ribosomal gene (rDNA) cluster. Here we show that recombinational repair of a DSB in rDNA in Saccharomyces cerevisiae involves the transient relocalization of the lesion to associate with the recombination machinery at an extranucleolar site. The nucleolar exclusion of Rad52 recombination foci entails Mre11 and Smc5–Smc6 complexes and depends on Rad52 SUMO (small ubiquitin-related modifier) modification. Remarkably, mutations that abrogate these activities result in the formation of Rad52 foci within the nucleolus and cause rDNA hyperrecombination and the excision of extrachromosomal rDNA circles. Our study also suggests a key role of sumoylation for nucleolar dynamics, perhaps in the compartmentalization of nuclear activities.
Article
Full-text available
Regulation of ribonucleotide reductase (RNR) is important for cell survival and genome integrity in the face of genotoxic stress. The Mec1/Rad53/Dun1 DNA damage response kinase cascade exhibits multifaceted controls over RNR activity including the regulation of the RNR inhibitor, Sml1. After DNA damage, Sml1 is degraded leading to the up-regulation of dNTP pools by RNR. Here, we probe the requirements for Sml1 degradation and identify several sites required for in vivo phosphorylation and degradation of Sml1 in response to DNA damage. Further, in a strain containing a mutation in Rnr1, rnr1-W688G, mutation of these sites in Sml1 causes lethality. Degradation of Sml1 is dependent on the 26S proteasome. We also show that degradation of phosphorylated Sml1 is dependent on the E2 ubiquitin-conjugating enzyme, Rad6, the E3 ubiquitin ligase, Ubr2, and the E2/E3-interacting protein, Mub1, which form a complex previously only implicated in the ubiquitylation of Rpn4.
Article
The recent explosion in the diversity of available fluorescent proteins (FPs) promises a wide variety of new tools for biological imaging. With no unified standard for assessing these tools, however, a researcher is faced with difficult questions. Which FPs are best for general use? Which are the brightest? What additional factors determine which are best for a given experiment? Although in many cases, a trial-and-error approach may still be necessary in determining the answers to these questions, a unified characterization of the best available FPs provides a useful guide in narrowing down the options.
Article
Homologous recombination is an important pathway for error-free repair of DNA lesions, such as single- and double-strand breaks, and for rescue of collapsed replication forks. Here, we describe protocols for live cell imaging of single-lesion recombination events in the yeast Saccharomyces cerevisiae using fluorescence microscopy.
Article
DPB11/TopBP1 is an essential evolutionarily conserved gene involved in initiation of DNA replication and checkpoint signaling. Here, we show that Saccharomyces cerevisiae Dpb11 forms nuclear foci that localize to sites of DNA damage in G1, S and G2 phase, a recruitment that is conserved for its homologue TopBP1 in Gallus gallus. Damage-induced Dpb11 foci are distinct from Sld3 replication initiation foci. Further, Dpb11 foci are dependent on the checkpoint proteins Mec3 (9-1-1 complex) and Rad24, and require the C-terminal domain of Dpb11. Dpb11 foci are independent of the checkpoint kinases Mec1 and Tel1, and of the checkpoint mediator Rad9. In a site-directed mutagenesis screen, we identify a separation-of-function mutant, dpb11-PF, that is sensitive to DSB-inducing agents yet remains proficient for DNA replication and the S-phase checkpoint at the permissive temperature. The dpb11-PF mutant displays altered rates of heteroallelic and direct-repeat recombination, sensitivity to DSB-inducing drugs as well as delayed kinetics of mating-type switching with a defect in the DNA synthesis step thus implicating Dpb11 in homologous recombination. We conclude that Dpb11/TopBP1 plays distinct roles in replication, checkpoint response and recombination processes, thereby contributing to chromosomal stability.
Article
A cell's ability to tolerate DNA damage is directly connected to the human development of diseases and cancer. To better understand the processes underlying mutagenesis, we studied the cell's reliance on the potentially error-prone translesion synthesis (TLS), and an error-free, template-switching pathway in Saccharomyces cerevisiae. The primary proteins mediating S. cerevisiae TLS are three DNA polymerases (Pols): Rev1, Pol ζ (Rev3/7), and Pol η (Rad30), all with human homologs. Rev1's noncatalytic role in recruiting other DNA polymerases is known to be important for TLS. However, the biological significance of Rev1's unusual conserved DNA polymerase activity, which inserts dC, is much less well understood. Here, we demonstrate that inactivating Rev1's DNA polymerase function sensitizes cells to both chronic and acute exposure to 4-nitroquinoline-1-oxide (4-NQO) but not to UV or cisplatin. Full Rev1-dependent resistance to 4-NQO, however, also requires the additional Rev1 functions. When error-free tolerance is disrupted through deletion of MMS2, Rev1's catalytic activity is more vital for 4-NQO resistance, possibly explaining why the biological significance of Rev1's catalytic activity has been elusive. In the presence or absence of Mms2-dependent error-free tolerance, the catalytic dead strain of Rev1 exhibits a lower 4-NQO-induced mutation frequency than wild type. Furthermore, Pol ζ, but not Pol η, also contributes to 4-NQO resistance. These results show that Rev1's catalytic activity is important in vivo when the cell has to cope with specific DNA lesions, such as N(2)-dG.
Article
We present the Flp-nick system, which allows introduction of a protein-bound nick at a single genomic site in Saccharomyces cerevisiae and thus mimics a stabilized topoisomerase I-DNA cleavage complex. We took advantage of a mutant Flp recombinase that can introduce a nick at a specific Flp recombinase recognition target site that has been integrated in the yeast genome. The genetic requirement for cells to cope with this insult is the same as for cells treated with camptothecin, which traps topoisomerase I-DNA cleavage complexes genome-wide. Hence, a single protein-bound nick is enough to kill cells if functional repair pathways are lacking. The Flp-nick system can be used to dissect repair, checkpoint and replication fork management pathways activated by a single genomic insult, and it allows the study of events at the damage site, which so far has been impossible to address.
Article
Homing endonucleases stimulate gene conversion by generating site-specific DNA double-strand breaks that are repaired by homologous recombination. These enzymes are potentially valuable tools for targeted gene correction and genome engineering. We have engineered a variant of the I-AniI homing endonuclease that nicks its cognate target site. This variant contains a mutation of a basic residue essential for proton transfer and solvent activation in one active site. The cleavage mechanism, DNA-binding affinity, and substrate specificity profile of the nickase are similar to the wild-type enzyme. I-AniI nickase stimulates targeted gene correction in human cells, in cis and in trans, at approximately 1/4 the efficiency of the wild-type enzyme. The development of sequence-specific nicking enzymes like the I-AniI nickase will facilitate comparative analyses of DNA repair and mutagenesis induced by single- or double-strand breaks.