Download Word

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cell culture wikipedia , lookup

Cellular differentiation wikipedia , lookup

List of types of proteins wikipedia , lookup

Mitosis wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Interferon wikipedia , lookup

Amitosis wikipedia , lookup

Transcript
1
Manuscript title: The putative U94 integrase is dispensable for human
2
herpesvirus 6 (HHV-6) chromosomal integration
3
4
Nina Wallaschek1, Annie Gravel2, Louis Flamand2,3, Benedikt B. Kaufer1,*
5
6
1Institut
7
14163 Berlin, Germany
8
2 Division
9
Quebec city, Quebec G1V 4G2, Canada
für Virologie, Freie Universität Berlin, Robert von Ostertag-Straße 7-13,
of Infectious Disease and Immunity, CHU de Québec Research Center,
10
3
11
Medicine, Université Laval, Quebec city, Québec,G1V 0A6 Canada
Department of microbiology, infectious disease and immunology, Faculty of
12
13
*Corresponding author:
14
Benedikt B. Kaufer, Institut für Virologie, Freie Universität Berlin, Robert von
15
Ostertag-Straße 7-13, 14163 Berlin, Germany.
16
Phone: +49-30-838-51936, FAX: +49-30-838-51847, e-mail: b.kaufer@fu-
17
berlin.de
18
19
Running title: Role of the U94 protein in HHV-6 integration
20
Key words: Human herpesvirus 6; integration; telomeres; latency; integrase; U94
21
Word count text: 1144
22
1
23
Abstract
24
Human herpesvirus 6 (HHV-6) can integrate its genome into the telomeres of
25
host chromosomes and is present in the germline of about 1 % of the human
26
population. HHV-6 encodes a putative integrase U94 that possesses all
27
molecular functions required for recombination like DNA-binding, ATPase,
28
helicase and nuclease activity and was hypothesized by many researchers to
29
facilitate integration, ever since the discovery of HHV-6 integration. However,
30
analysis of U94 in the virus context was hampered by the lack of reverse-genetic
31
systems and efficient integration assays. Here, we addressed the role of U94 and
32
the cellular recombinase Rad51 in HHV-6 integration. Surprisingly, we could
33
demonstrate that HHV-6 efficiently integrated in the absence of U94 using a new
34
quantitative integration assay. Additional inhibition of the cellular recombinase
35
Rad51 only had a minor impact on virus integration. Our results shed light on this
36
complex integration mechanism that includes factors beyond U94 and Rad51.
2
37
38
Text
Human
herpesvirus
-6A
and
-6B
(HHV-6)
are
closely
related
39
betaherpesviruses that infect humans early in life (Salahuddin et al., 1986). HHV-
40
6B is the causative agent of roseola infantum, a febrile illness in infants, while
41
disease associations of HHV-6A are not well characterized (Yamanishi et al.,
42
1988). Both viruses have previously been shown to integrate its genetic material
43
into telomeres of human chromosomes (Arbuckle et al., 2010, Arbuckle et al.,
44
2013). This integration mechanism also allows vertical transmission of the virus
45
via the germline, resulting in individuals that harbor the integrated virus in every
46
single cell of their body (Daibata et al., 1998, Daibata et al., 1999, Mori et al.,
47
2009, Tanaka-Taya et al., 2004, Kuhl et al., 2014, Osterrieder et al., 2014). This
48
condition is termed inherited chromosomally integrated HHV-6 (iciHHV-6) and is
49
present in about 1 % of the human population (Pellett et al., 2012). The biological
50
consequences of this condition are not well understood yet. A recent report
51
indicates that iciHHV-6+ subjects are at greater risks of developing angina
52
(Gravel et al., 2015). The molecular mechanism and the factors involved in HHV-
53
6 integration remain completely unknown.
54
HHV-6 encodes the U94 gene that contains all conserved domains of the
55
Rep68 integrase of Adeno-associated virus 2 (AAV-2) (Arbuckle and Medveczky,
56
2011, Thomson et al., 1991). Expression of U94 restores replication of a Rep-
57
deficient AAV-2, suggesting that both proteins have similar functions (Thomson
58
et al., 1994). U94 of both HHV-6A and HHV-6B has been shown to possess
59
single-strand and double-strand DNA binding activity with a preference for
3
60
TTAGGG repeats, as present in human telomeres. In addition, U94 has ATPase,
61
helicase and 3’-5’ exonuclease activity against 3’ recessive ends in vitro (Trempe
62
et al., 2015). Until now, the role of U94 in virus replication and integration could
63
not be determined due to the lack of reverse-genetic systems and efficient
64
integration assays that would allow quantification of virus integration.
65
To investigate the role of U94 in HHV-6 integration, we deleted the entire
66
U94 gene (ΔU94) in pHHV-6A (wt), a recently generated infectious bacterial
67
artificial chromosome (BAC) clone of HHV-6A (strain U1102), using two-step
68
Red-mediated mutagenesis (Fig. 1a) as published previously (Tang et al., 2010,
69
Tischer et al., 2006, Tischer and Kaufer, 2012). In addition, we generated a
70
revertant virus in which we restored U94 to ensure that no secondary mutations
71
occurred during the mutagenesis process. Primers used for the mutagenesis are
72
listed in Table S1. Recombinant BAC clones were confirmed by RFLP, DNA
73
sequencing and Southern blotting using a specific digoxigenin (DIG)-labeled
74
probe for U94 (Fig. 1b) as described previously (Kaufer et al., 2011). The
75
recombinant viruses were subsequently reconstituted by nucleofection of JJHan
76
cells with the HHV-6A BAC DNA as described previously (Nukui et al., 2015).
77
Cell-free virus stocks were generated by concentrating the supernatant of highly
78
infected JJHan cells and titrated by analyzing the genome copies in newly
79
infected cells by qPCR. Multi-step growth kinetics (Oyaizu et al., 2012, Nukui et
80
al., 2015) revealed that the ΔU94 mutant has a significant growth defect
81
compared to the wt and revertant virus (Fig. 1c), suggesting that the impaired
82
HHV-6A replication is either due to the abrogation of U94 expression or effects
4
83
on the neighboring genes such as U95 caused by the deletion of the U94
84
sequences.
85
To determine the integration efficiency of recombinant HHV-6A viruses,
86
we used our recently established integration assay (Gravel et al, submitted).
87
Briefly, U2OS cells were infected with wt, ΔU94 and ΔU94rev that express GFP
88
under the control of the HCMV major immediate early promoter. Infected GFP-
89
positive cells were isolated 36 h post infection using a FACS AriaIII cell sorter
90
(BD, San Jose), providing a pure infected cell population for our integration
91
assays. To determine the integration efficiency of respective viruses, we
92
performed FISH analyses on the infected U2OS cultures at day 14 post sorting
93
as described previously (Kaufer et al., 2011, Kaufer, 2013). Despite the growth
94
defect of ΔU94 during lytic replication, the integration frequency of ΔU94 in
95
metaphase chromosomes was comparable to wt and revertant virus (Fig. 2a-b);
96
for this purpose 90 metaphases for each virus were imaged and the proportion
97
(%) determined that showed a specific signal for HHV-6 at the ends of
98
chromosomes in two chromatids. To confirm that this is not a metaphase specific
99
effect, we analyzed interphase nuclei for the presence of the virus genome.
100
ΔU94 was detected in comparable proportion of nuclei (Fig. 2c). These findings
101
indicate that U94 is not essential for HHV-6A integration. Furthermore, we
102
performed qPCR analyses at day 0 and 14 post sorting to determine if the
103
maintenance of the HHV-6A genome was altered upon abrogation of U94. Viral
104
genome copies were quantified by qPCR relative to the cellular genome, using
105
primers and probes specific for HHV-6A U86 and the cellular ß2M gene,
5
106
respectively (Table S1). The HHV-6A genome was maintained in ΔU94 infected
107
cells at comparable levels as wt and revertant viruses (Fig. 2d). Furthermore,
108
clonal cell lines for wt, ΔU94 and ΔU94rev were generated to confirm the
109
integration into host telomeres (Figure 3).
110
To exclude that the alternative lengthening of telomeres (ALT) activity
111
present in U2OS cells can compensate for the absence of U94, we confirmed our
112
observations in ALT negative 293T cells (Figure S1) and JJHan cells (data not
113
shown). Furthermore, no integration defect was observed in a HHV-6A mutant
114
virus in which U94 expression was abrogated by insertion of multiple stop codons
115
within the open reading frame (Figure S2), while viruses that lacked the viral
116
telomere sequences had a severe integration defect (Wallaschek et al.,
117
submitted). Taken together, our data demonstrates that U94 is not essential for
118
HHV-6A integration in U2OS, 293T and JJHan cells.
119
Next, we investigated if the cellular recombinase Rad51 that is essential
120
for homologous recombination in mitotic cells (San Filippo et al., 2008) is
121
involved in the integration of HHV-6A and could facilitate integration in the
122
absence of U94. Therefore, we performed in vitro integration assays with wt and
123
ΔU94 mutant viruses in the presence or absence of the specific Rad51 inhibitor
124
RI-1. We used 5 µM of RI-1 as this concentration was previously shown to
125
efficiently inhibit Rad51 (Budke et al., 2012), while no cytotoxic effects were
126
detectable (Figure S3). Both wt and the ΔU94 mutant were still able to integrate
127
into U2OS cells. A slightly decreased frequency in metaphases and interphases
128
was observed (Fig. 2e and f), but our data suggest that Rad51 does not play a
6
129
major role in the integration of HHV-6A in U2OS cells. Comparable results were
130
obtained with the commercial Rad51 inhibitor B02 (data not shown). Similarly,
131
qPCR analyses revealed a minor reduction in HHV-6A genome copies in Rad51
132
treated cells (Fig. 2g). Taken together, our data demonstrates that both U94, the
133
putative viral integrase, and Rad51, the cellular recombinase, are dispensable for
134
the integration of HHV-6A into host chromosomes in the two tested cell lines,
135
suggesting that other viral or cellular recombinases can complement their
136
functions. Most likely, several redundant pathways ensure that HHV-6 integration
137
can occur. Besides homologous recombination (HR), also single strand
138
annealing (SSA) is a possible mechanism that could accomplish virus integration.
139
In this scenario, cellular factors such as Rad52 or viral factors including U41 and
140
U70, the homologs of HSV-1 UL12 and ICP8, which were shown to mediate
141
strand exchange (Reuven et al., 2003, Schumacher et al., 2012), could also
142
facilitate HHV-6 integration and are targets of future studies. In addition, we will
143
established a quantitative integration assay for primary cells to assess the role of
144
U94 in cells ex vivo.
145
146
147
Acknowledgements
148
We are grateful to Yasuko Mori (Kobe University, Japan) for providing the HHV-
149
6A BAC and Ann Reum for her technical assistance. This study was supported
150
by the DFG grant KA 3492/1-1 and the ERC starting grant StG-677673 awarded
151
to BK.
152
7
153
References
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
197
198
ARBUCKLE, J. H., MEDVECZKY, M. M., LUKA, J., HADLEY, S. H., LUEGMAYR,
A., ABLASHI, D., LUND, T. C., TOLAR, J., DE MEIRLEIR, K., MONTOYA,
J. G., KOMAROFF, A. L., AMBROS, P. F. & MEDVECZKY, P. G. 2010. The
latent human herpesvirus-6A genome specifically integrates in telomeres of
human chromosomes in vivo and in vitro. Proc Natl Acad Sci U S A, 107, 5563-8.
ARBUCKLE, J. H. & MEDVECZKY, P. G. 2011. The molecular biology of human
herpesvirus-6 latency and telomere integration. Microbes Infect, 13, 731-41.
ARBUCKLE, J. H., PANTRY, S. N., MEDVECZKY, M. M., PRICHETT, J., LOOMIS,
K. S., ABLASHI, D. & MEDVECZKY, P. G. 2013. Mapping the telomere
integrated genome of human herpesvirus 6A and 6B. Virology, 442, 3-11.
BUDKE, B., LOGAN, H. L., KALIN, J. H., ZELIVIANSKAIA, A. S., CAMERON
MCGUIRE, W., MILLER, L. L., STARK, J. M., KOZIKOWSKI, A. P.,
BISHOP, D. K. & CONNELL, P. P. 2012. RI-1: a chemical inhibitor of RAD51
that disrupts homologous recombination in human cells. Nucleic Acids Res, 40,
7347-57.
DAIBATA, M., TAGUCHI, T., NEMOTO, Y., TAGUCHI, H. & MIYOSHI, I. 1999.
Inheritance of chromosomally integrated human herpesvirus 6 DNA. Blood, 94,
1545-9.
DAIBATA, M., TAGUCHI, T., SAWADA, T., TAGUCHI, H. & MIYOSHI, I. 1998.
Chromosomal transmission of human herpesvirus 6 DNA in acute lymphoblastic
leukaemia. Lancet, 352, 543-4.
GRAVEL, A., DUBUC, I., MORISSETTE, G., SEDLAK, R. H., JEROME, K. R. &
FLAMAND, L. 2015. Inherited chromosomally integrated human herpesvirus 6
as a predisposing risk factor for the development of angina pectoris. Proc Natl
Acad Sci U S A, 112, 8058-63.
KAUFER, B. B. 2013. Detection of integrated herpesvirus genomes by fluorescence in
situ hybridization (FISH). Methods Mol Biol, 601-6_10.
KAUFER, B. B., JAROSINSKI, K. W. & OSTERRIEDER, N. 2011. Herpesvirus
telomeric repeats facilitate genomic integration into host telomeres and
mobilization of viral DNA during reactivation. J Exp Med, 208, 605-15.
KUHL, U., LASSNER, D., WALLASCHEK, N., GROSS, U. M., KRUEGER, G. R.,
SEEBERG, B., KAUFER, B. B., ESCHER, F., POLLER, W. & SCHULTHEISS,
H. P. 2014. Chromosomally integrated human herpesvirus 6 in heart failure:
prevalence and treatment. Eur J Heart Fail.
MORI, T., TANAKA-TAYA, K., SATOH, H., AISA, Y., YAMAZAKI, R., KATO, J.,
IKEDA, Y. & OKAMOTO, S. 2009. Transmission of chromosomally integrated
human herpesvirsus 6 (HHV-6) variant A from a parent to children leading to
misdiagnosis of active HHV-6 infection. Transpl Infect Dis, 11, 503-6.
NUKUI, M., MORI, Y. & MURPHY, E. A. 2015. A human herpesvirus 6A-encoded
microRNA: role in viral lytic replication. J Virol, 89, 2615-27.
OSTERRIEDER, N., WALLASCHEK, N. & KAUFER, B. B. 2014. Herpesvirus
Genome Integration into Telomeric Repeats of Host Cell Chromosomes. Annual
Review of Virology, 1, 215-235.
OYAIZU, H., TANG, H., OTA, M., TAKENAKA, N., OZONO, K., YAMANISHI, K. &
MORI, Y. 2012. Complementation of the function of glycoprotein H of human
8
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
215
216
217
218
219
220
221
222
223
224
225
226
227
228
229
230
231
232
233
234
235
236
237
238
239
240
241
242
243
herpesvirus 6 variant A by glycoprotein H of variant B in the virus life cycle. J
Virol, 86, 8492-8.
PELLETT, P. E., ABLASHI, D. V., AMBROS, P. F., AGUT, H., CASERTA, M. T.,
DESCAMPS, V., FLAMAND, L., GAUTHERET-DEJEAN, A., HALL, C. B.,
KAMBLE, R. T., KUEHL, U., LASSNER, D., LAUTENSCHLAGER, I.,
LOOMIS, K. S., LUPPI, M., LUSSO, P., MEDVECZKY, P. G., MONTOYA, J.
G., MORI, Y., OGATA, M., PRITCHETT, J. C., ROGEZ, S., SETO, E., WARD,
K. N., YOSHIKAWA, T. & RAZONABLE, R. R. 2012. Chromosomally
integrated human herpesvirus 6: questions and answers. Rev Med Virol, 22, 14455.
REUVEN, N. B., STAIRE, A. E., MYERS, R. S. & WELLER, S. K. 2003. The herpes
simplex virus type 1 alkaline nuclease and single-stranded DNA binding protein
mediate strand exchange in vitro. J Virol, 77, 7425-7433.
SALAHUDDIN, S. Z., ABLASHI, D. V., MARKHAM, P. D., JOSEPHS, S. F.,
STURZENEGGER, S., KAPLAN, M., HALLIGAN, G., BIBERFELD, P.,
WONG-STAAL, F., KRAMARSKY, B. & ET AL. 1986. Isolation of a new
virus, HBLV, in patients with lymphoproliferative disorders. Science, 234, 596601.
SAN FILIPPO, J., SUNG, P. & KLEIN, H. 2008. Mechanism of eukaryotic homologous
recombination. Annu Rev Biochem, 77, 229-57.
SCHUMACHER, A. J., MOHNI, K. N., KAN, Y., HENDRICKSON, E. A., STARK, J.
M. & WELLER, S. K. 2012. The HSV-1 exonuclease, UL12, stimulates
recombination by a single strand annealing mechanism. PLoS Pathog, 8,
e1002862.
TANAKA-TAYA, K., SASHIHARA, J., KURAHASHI, H., AMO, K., MIYAGAWA,
H., KONDO, K., OKADA, S. & YAMANISHI, K. 2004. Human herpesvirus 6
(HHV-6) is transmitted from parent to child in an integrated form and
characterization of cases with chromosomally integrated HHV-6 DNA. J Med
Virol, 73, 465-73.
TANG, H., KAWABATA, A., YOSHIDA, M., OYAIZU, H., MAEKI, T.,
YAMANISHI, K. & MORI, Y. 2010. Human herpesvirus 6 encoded glycoprotein
Q1 gene is essential for virus growth. Virology, 407, 360-7.
THOMSON, B. J., EFSTATHIOU, S. & HONESS, R. W. 1991. Acquisition of the
human adeno-associated virus type-2 rep gene by human herpesvirus type-6.
Nature, 351, 78-80.
THOMSON, B. J., WEINDLER, F. W., GRAY, D., SCHWAAB, V. & HEILBRONN, R.
1994. Human herpesvirus 6 (HHV-6) is a helper virus for adeno-associated virus
type 2 (AAV-2) and the AAV-2 rep gene homologue in HHV-6 can mediate
AAV-2 DNA replication and regulate gene expression. Virology, 204, 304-11.
TISCHER, B. K. & KAUFER, B. B. 2012. Viral bacterial artificial chromosomes:
generation, mutagenesis, and removal of mini-F sequences. J Biomed Biotechnol,
472537, 23.
TISCHER, B. K., VON EINEM, J., KAUFER, B. & OSTERRIEDER, N. 2006. Two-step
red-mediated recombination for versatile high-efficiency markerless DNA
manipulation in Escherichia coli. Biotechniques, 40, 191-7.
9
244
245
246
247
248
249
250
251
TREMPE, F., GRAVEL, A., DUBUC, I., WALLASCHEK, N., COLLIN, V., GILBERTGIRARD, S., MORISSETTE, G., KAUFER, B. B. & FLAMAND, L. 2015.
Characterization of human herpesvirus 6A/B U94 as ATPase, helicase,
exonuclease and DNA-binding proteins. Nucleic Acids Res, 43, 6084-98.
YAMANISHI, K., OKUNO, T., SHIRAKI, K., TAKAHASHI, M., KONDO, T.,
ASANO, Y. & KURATA, T. 1988. Identification of human herpesvirus-6 as a
causal agent for exanthem subitum. Lancet, 1, 1065-7.
10
252
Figure legends
253
Figure 1: Generation and characterization of the ΔU94 mutant and ΔU94
254
revertant. (a) Schematic representation of the HHV-6A wt, the ΔU94 mutant and
255
the ΔU94rev. (b) Southern blot analysis after HindIII digestion of the indicated
256
BAC clones using a DIG-labeled U94 probe. (c) Growth kinetics comparing
257
replication properties of HHV-6A wt, ΔU94 mutant and ΔU94rev virus in JJHan
258
cells. HHV-6A genome copy numbers were determined by qPCR. Copy numbers
259
per 1x 106 cells are shown as means of three independent experiments with
260
standard errors. Significant differences between wt and ΔU94 as well as ΔU94
261
and ΔU94rev (Mann-Whitney U-test, p < 0.05 or p < 0.01) are indicated with
262
asterisks (* or **). dpi: days post infection.
263
Figure 2: Integration efficiency and genome maintenance of the ΔU94
264
mutant in the U2OS integration system. (a) Representative metaphase images
265
of HHV-6A wt, ΔU94 and ΔU94rev virus in U2OS cells at d14 day post sorting.
266
Cells were fixed, metaphase spreads were prepared, hybridized with a HHV-6A
267
specific DIG-labeled probe, washed and incubated with a secondary anti-DIG-
268
FITC antibody, DAPI was used to visualize the nuclei and chromosomes. Blue:
269
DNA, green: HHV-6A genome. Scale bar corresponds to 10µm. (b and e)
270
Integration frequency of indicated viruses in the absence (b) and presence of the
271
specific Rad51 inhibitor RI-1 (e) was quantified by analyzing the integration
272
status of at least 60 metaphases. Results are shown as means of two (e) or three
273
(b) independent experiments with standard errors. (c and f) >200 interphase
274
nuclei of cells infected in the absence (c) and presence of the specific Rad51
275
inhibitor RI-1 (f) with indicated viruses were examined for the presence of HHV-
276
6A. Results are shown as means of two (f) or three (c) independent experiments
277
with standard errors. (d and g) Maintenance of the HHV-6A genome in the
278
absence (d) and presence of the specific Rad51 inhibitor RI-1 (g) was
279
determined by qPCR analysis at d0 and d14 post sort. Copy numbers per 1x 10 6
280
cells are shown as means of three independent experiments with standard
11
281
errors. Significant differences between ΔU94 and ΔU94 + RI-1 (Mann-Whitney U-
282
test, p < 0.05) are indicated with an asterisk (*).
283
Figure 3: Generation of clonal U2OS cell lines containing wt, ΔU94 and
284
ΔU94rev virus. The HHV-6A genome was detected using a specific DIG-labeled
285
probe by FISH (green) and nuclei and chromosomes were visualized using DAPI
286
(blue). Representative images of clonal U2OS cell lines for wt, ΔU94 and
287
ΔU94rev are shown. Metaphases are shown on the left, interphase nuclei on the
288
right. Scale bar corresponds to 10µm.
289
Figure S1: Genome maintenance and integration of the ΔU94 mutant in
290
293T cells. (a) Maintenance of the HHV-6A genome was determined by qPCR
291
analysis at d0 and d14 post sort. Copy numbers per 1x 10 6 cells are shown as
292
means of two independent experiments with standard errors. (b) Representative
293
metaphase images of HHV-6A wt and ΔU94 virus in 293T cells at d14 day post
294
sorting. Cells were fixed, metaphase spreads were prepared, hybridized with a
295
HHV-6A specific DIG-labeled probe, washed and incubated with a secondary
296
anti-DIG-FITC antibody,
297
chromosomes. Blue: DNA, green: HHV-6A genome.
298
Figure S2: Integration efficiency and genome maintenance of the U94stop
299
mutant in the U2OS integration system. (a) Integration frequency of indicated
300
viruses was quantified by analyzing the integration status of 90 metaphases.
301
Results are shown as means of three independent experiments with standard
302
errors. (b) 300 interphase nuclei of cells infected with indicated viruses were
303
examined for the presence of HHV-6A. Results are shown as means of three
304
independent experiments with standard errors. (c) Maintenance of the HHV-6A
305
genome was determined by qPCR analysis at d0 and d14 post sort. Copy
306
numbers per 1x 106 cells are shown as means of three independent experiments
307
with standard errors.
DAPI
was used
12
to
visualize
the
nuclei and
308
Figure S3: Killing curve for determination of the optimal Rad51 inhibitor
309
concentration. U2OS cells were treated with increasing concentrations of the
310
Rad51 inhibitor RI-1 and cell viability was checked by microscopy.
13