ArticlePDF Available

Expression strategy of Aedes albopictus densovirus (AalDNV).

American Society for Microbiology
Journal of Virology
Authors:
  • McGill AIDS Centre, Lady Davis Institute, Jewish General Hospital

Abstract and Figures

The transcription map of the Aedes albopictus densovirus (AalDNV) brevidensovirus was identified by Northern blotting, rapid amplification of cDNA ends (RACE) analysis, and RNase protection assays. AalDNV produced mRNAs of 3,359 (NS1), 3,345 (NS2), and 1,246 (VP) nucleotides. The two overlapping P7/7.4 NS promoters employed closely located alternate transcription initiation sites, positioned at either side of the NS1 initiation codon. All NS mRNAs coterminated with VP mRNA. All promoters, explored using luciferase assays, were functional in insect and human cell lines.
Content may be subject to copyright.
Published Ahead of Print 26 June 2013.
2013, 87(17):9928. DOI: 10.1128/JVI.01259-13. J. Virol.
Peter Tijssen
Perreault, Hiroko Shike, Jozsef Szelei, Max Bergoin and
Hanh T. Pham, Françoise-Xavière Jousset, Jonathan
Densovirus
Expression Strategy of Aedes albopictus
http://jvi.asm.org/content/87/17/9928
Updated information and services can be found at:
These include:
REFERENCES
http://jvi.asm.org/content/87/17/9928#ref-list-1
This article cites 15 articles, 7 of which can be accessed free at:
CONTENT ALERTS
more»articles cite this article),
Receive: RSS Feeds, eTOCs, free email alerts (when new
http://journals.asm.org/site/misc/reprints.xhtmlInformation about commercial reprint orders:
http://journals.asm.org/site/subscriptions/To subscribe to to another ASM Journal go to:
on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from
Expression Strategy of Aedes albopictus Densovirus
Hanh T. Pham,
a
Françoise-Xavière Jousset,
b
Jonathan Perreault,
a
Hiroko Shike,
c
Jozsef Szelei,
a
Max Bergoin,
a
Peter Tijssen
a
INRS-Institut Armand-Frappier, Laval, Québec, Canada
a
; Laboratoire de Pathologie Comparée, Université Montpellier II, Montpellier, France
b
; Division of Clinical Pathology,
Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, Pennsylvania, USA
c
The transcription map of the Aedes albopictus densovirus (AalDNV) brevidensovirus was identified by Northern blotting, rapid
amplification of cDNA ends (RACE) analysis, and RNase protection assays. AalDNV produced mRNAs of 3,359 (NS1), 3,345
(NS2), and 1,246 (VP) nucleotides. The two overlapping P7/7.4 NS promoters employed closely located alternate transcription
initiation sites, positioned at either side of the NS1 initiation codon. All NS mRNAs coterminated with VP mRNA. All promot-
ers, explored using luciferase assays, were functional in insect and human cell lines.
I
nvertebrate parvoviruses (densoviruses [DNVs]) are subdivided
into four genera: Densovirus, Pefudensovirus, Iteravirus, and
Brevidensovirus (1–3). Brevidensoviruses have a 4.1-kb single-
stranded DNA (ssDNA) genome with three open reading frames
(ORFs) on the same strand (about 790 [NS1], 360 [NS2], and 350
[VP] amino acids). The genome of Aedes albopictus densovirus
(AalDNV) (GenBank accession no. NC_004285) has terminal, T-
shaped hairpins (4). Brevidensoviruses were isolated from medi-
cally important mosquito vectors, such as Aedes albopictus
(AalDNV) (4), Aedes aegypti (AaeDNV) (5), and Anopheles gam-
biae (AgDNV) (6). AalDNV was isolated from Aedes albopictus
C6/36 cells (7) but is infectious for Aedes aegypti larvae (8, 9).
AaeDNV expression has been studied to some extent (10, 11).
ORF
NS1
of AaeDNV was reported to have a 57-amino-acid N-ter-
minal extension compared to that of AalDNV (4, 5). -Galactosi-
dase fusion proteins with the three ORFs were enzymatically ac-
tive, except for NS1 (12) unless NS1--gal was constructed
downstream of the corresponding AalDNV AUG
NS1
(11). Primer
extension demonstrated that AaeDNV VP transcription started at
nucleotide (nt) 2402 (10). Here, AalDNV transcription was ana-
lyzed by Northern hybridization, 5= and 3= rapid amplification of
cDNA ends (RACE), amplicon mapping, and RNase protection
assays (RPAs). AalDNV promoter activities were also determined.
AalDNV transcripts were obtained after transfection of
pCR2.1-AalDNV (containing the AalDNV genome of 4,176 nt [4]
between its EcoRI sites), using Lipofectamine (Invitrogen proto-
col), in permissive C6/36 cells (7) grown in RPMI medium sup-
plemented with 5% fetal bovine serum (FBS). After 48 h, RNA was
isolated using the Stratagene Absolutely RNA miniprep kit.
Northern blotting.
32
P-labeled RNA probes for Northern
blots targeted overlapping domains of the NS1 and NS2 ORFs or
VP ORF. PCR-amplified products (primers in Table 1) were tran-
scribed in vitro with [-
32
P]UTP and T7 RNA polymerase (NEB)
(13). Northern blots using 10 g of total RNA (13, 14) revealed
transcript sizes of 3.4 and 1.3 kb with the VP-specific probe and a
3.4-kb transcript with the NS-specific probe (Fig. 1A).
RACE experiments. The FirstChoice RLM RACE kit (Invitro-
gen) was used to identify the 5= starts and 3= ends of the polyadenyl-
ated transcripts. The 5= adapter primer (IP) and the 3=-anchored
primer (AP) (in the kit) were used in PCR with AalDNV-specific
primers (Table 1), as shown in Fig. 1B. Amplicons obtained with
A626R/IP (325 bp), A497R/IP (200 bp), A2740R/IP (350 bp),
A2540R/IP (150 bp), A3162F/AP (600 bp), and A3461F (300
bp) (data not shown) were sequenced. Sequencing revealed that NS1
transcription started at nt 329 (AGTA), 6 nt upstream of AUG
NS1
,
and that VP transcription started at nt 2441 (CAGTCG), 158 nt up-
stream of AUG
VP
(Fig. 1B) (sequence data not shown). Sequencing of
the 300- and 600-bp amplicons showed a transcription termination
position at 3680, 18 nt downstream of the polyadenylation signal at
position 3662 (Fig. 1C) (sequence data not shown).
As is common for other densoviruses, NS2 could be translated
from nt 411 by leaky scanning (13) on the NS1 transcript. The
short untranscribed region of NS1 (UTR
NS1
), in spite of a consen-
sus AnnAUGG sequence for NS1 initiation, could favor it (13). A
canonical initiator Inr
NS2
sequence (CAGT) is located at nt 342.
After TfiI digestion (NEB) at nt 338 of the A831R/IP PCR product,
followed by a nested PCR (A626R/IP or A497R/IP) for specific
amplification of putative NS2 transcripts, bands of 330 and 200 nt
were produced (data not shown). Their sequencing revealed that
NS2 transcription started at nt 343 (sequence data not shown).
Thus, NS1 and NS2 AalDNV transcription starts were separated
by 14 nt on either side of AUG
NS1
.
Amplicon mapping. In order to confirm that NS and VP tran-
scripts all coterminated at position 3680, primer A2380F (up-
stream of the VP transcript) and two reverse primers, A3650R
(upstream) and A3744R (downstream), of the VP transcript end
were designed for reverse transcription (RT)-PCR amplification
(Table 1). Only a band of about 1,300 nt was obtained using the
A2380F/A3650R set of primers for NS transcripts (Fig. 1C).
RNase protection assays. RPAs were employed to confirm
NS1, NS2, and VP transcription starts. RPA probes for NS and VP
transcripts were prepared by PCR (Table 1; Fig. 2A), and in vitro
transcription as described for Northern blot probes. For positive
controls, a sense RNA was generated by in vitro transcription and
RPA was performed in parallel: for NS, two positive-control RNAs
spanning from nt 329 to 439 (predicted for NS1) and nt 343 to 439
(predicted for NS2) were used, and for VP one from nt 2441 to
2542 was used. The probes and control RNA were purified from
polyacrylamide gel and used in the RPA III kit (Invitrogen). Size
markers were generated according to the 5= RACE results. The NS
Received 9 May 2013 Accepted 18 June 2013
Published ahead of print 26 June 2013
Address correspondence to Peter Tijssen, peter.tijssen@iaf.inrs.ca.
Copyright © 2013, American Society for Microbiology. All Rights Reserved.
doi:10.1128/JVI.01259-13
9928 jvi.asm.org Journal of Virology p. 9928 –9932 September 2013 Volume 87 Number 17
on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from
probe protected the expected 96 and 110 nt of the NS transcript 5=
ends (Fig. 2B), confirming NS1 and NS2 mRNA start positions.
Similarly, the VP probe confirmed the VP transcript 5= extremity
by protecting 101 nt (Fig. 2B). A protected band at 219 nt, slightly
smaller than the entire probe (236 nt, including 17 nt of unspecific
sequence), showed the coexistence of NS transcripts.
Promoter activity. NS (P7/7.4) and VP (P60) promoter re-
gions were amplified by PCR (primers in Table 1) and cloned into
TABLE 1 Primers used for Northern blot probes, RACE, amplicon mapping, and RPAs
Primer name
a
Sequence Purpose
A639F GCTCCAGAGCCTCTGAACAGCTTG NS probe, Northern blot
A1345R (T7 sequence) TAATACGACTCACTATAGGGGTTCTGACTCTTGTGCTGTTTC
NS probe, Northern blot
A3106F CTAGAAACAGTTGCAGCAACCGGAC VP probe, Northern blot
A3509R (T7 sequence) TAATACGACTCACTATAGGCGTACTTGATATCTGAATTTCATG
VP probe, Northern blot
A3372F AACTACAACATATGCCACGTCAG 3= RACE
A3461F ACAAGTTCCAGACGAAACAGG 3= RACE
A497R GTTCGTAATTGTTGGCATTCCT 5= RACE of NS
A626R GTGGGTAGATGTTATCAACGG 5= RACE of NS
A831R CTTGCCTGTGACCCGTTATTATCC 5= RACE of NS
A2540R GTGCGTTGTCTTCTTCTTCTATC 5= RACE of VP
A2740R GACCAAACATTACGGAAATGG 5= RACE of VP
A3126F CGGACCATTAGCACAACAAAC 3= RACE
A2380F GAGTATACAACACAGAGAAG Amplicon mapping
A3650R TCATAA GGCATACATGCTAC Amplicon mapping
A3744R TCTGTCGTGGACATTATCAG Amplicon mapping
A272F (UTS) GCGATGAATGAACACTGAATCCACCACCACATGATCC
RPA NS probe
A329F (T7 sequence) TAATACGACTCACTATAGGAGTAGTATGGAATCAG RPA-positive control for NS1
A343F (T7 sequence) TAATACGACTCACTATAGGGTCTGCAGTGAACATTCG
RPA-positive control for NS2
A439R TCTCCTCCTGGATTTACACTG RPA-positive control for NS1 and NS2
A439R (T7 sequence) TAATACGACTCACTATAGGTCTCCTCCTGGATTTACACTG
RPA NS probe
A2323F (UTS) GCGATGAATGAACACTGGCATATGAACGAAACCTCAC
RPA VP probe
A2441F (T7 sequence) TAATACGACTCACTATAGGAGTCGGCCACCAGGTCTTGTAG
RPA-positive control for VP
A2542R ATGTGCGTTGTCTTCTTCTTC RPA-positive control for VP
A2542R (T7 sequence) TAATACGACTCACTATAGGATGTGCGTTGTCTTCTTCTTC RPA VP probe
A148F TCCAATTGGAACACACGGAC P7/7.4 promoter for AalDNV
A333R CTACTGACTCTCCCTTC P7/7.4 promoter for AalDNV
A2431F CAAACTCATCAGTCGGCCAC P60 promoter for AalDNV
A2597R CCTCTGCTTCTTCTTTTGC P60 promoter for AalDNV
a
In the primer names, “UTS” represents the unspecific target sequence and “R” and “F” represent sense and antisense, respectively. The numbers in the primer names indicate the
5= end of the primer sequence in AalDNV. The underlined sequences represent the UTS or T7 sequences.
FIG 1 (A) Northern blot analysis of AalDNV transcription 48 h after transfection with vectors with virus-specific inserts (lanes 1) or without inserts (lanes 2).
The VP probe also detected bands corresponding to the size of NS transcript indicating that VP and NS transcripts may coterminate. (B) Strategy of 5= and 3=
RACE. Above the ORFs are the specific primers that were designed, and below are the ORFs and IP and AP primers from the Invitrogen kit (indicated with
arrows). We took advantage of the TfiI restriction site between the putative Inr of NS1 and NS2 to distinguish between these transcripts. (After digestion, only
amplicons from the second Inr would be obtained.) (C) Confirmation of termini of NS transcripts of AalDNV. The diagrams represent transcript map results by
5= and 3= RACE. Forward primers that do not recognize VP transcripts and reverse primers, both upstream and downstream of the VP transcript end (3= RACE),
were used in a PCR (indicated in the diagrams). As expected, only the inboard reverse primer (A3650R in lane 1) gave a product. M, markers; lane 2,
A2380F/A3744R.
Expression of the AalDNV Brevidensovirus
September 2013 Volume 87 Number 17 jvi.asm.org 9929
on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from
EcoRV in pGL4.20, upstream of the luciferase gene (Promega;
GenBank accession no. DQ188840) to estimate their functional-
ity. Those with the reverse promoter orientation, shown by se-
quencing, served as controls. The Mythimna loreyi (MlDNV) NS
promoter (an insect virus replicating in LD652 cells) (15) and the
simian virus 40 (SV40) promoter were also cloned into pGL4.20
vectors to serve as positive controls. Transfection of LD652 using
DOTAP (Roche) and HeLa cells using Lipofectamine (Invitrogen)
in 24-well plates was performed according to the suppliers’ in-
structions. Cells were harvested 40 h posttransfection and resus-
pended in 100 l of Bright-Glo lysis buffer (Promega), and rela-
tive luciferase activity was determined according to Promega’s
instructions. The AalDNV P7/7.4 and P60 promoters were func-
tional both in insect cells and in human cells, in contrast to con-
structs lacking a promoter or with promoters in the reverse orien-
tation, whereas the SV40 promoter preferred HeLa cells and the
MlDNV promoter preferred insect cells (Fig. 2C).
(i) Promoter 7/7.4 elements. The RACE experiments and
RPA (Fig. 2) revealed the location of Inr1 and Inr2 at either side
of the NS1 protein initiation codon (Fig. 3C). Consequently,
the NS1 transcript could potentially be translated into both
NS1 and NS2 proteins, whereas the NS2 transcript could only
code for the NS2 protein. Different constructs, leaving Inr1 and
its upstream elements intact (for NS1 transcription) but with
mutations in the NS2 promoter and NS1/2 translation ele-
ments, were made with a luciferase reporter gene (Fig. 3B and
C). Constructs containing the intact luciferase AUG (F1 to F7)
served as a positive control for transcription, whereas those
lacking the luciferase AUG (F1 to F7) showed the impact of
the various mutations.
P7/7.4 consisted of three segments (Fig. 3B). The KpnI-SacI
segment I contained upstream promoter elements, the SacI-PstI
segment II included both TATA boxes, ATG
NS1
, and both Inrs
and, finally, the PstI-HindIII segment III (about 20 nt for the
NS1 ORF and about 70 nt for the NS2 ORF) contained the
downstream promoter elements (DPE) and ATG
NS2
. These
segments were connected in pBluescript (PCR with primers
containing appropriate restriction sites and pCR2.1-AalDNV
template) and were transferred as a whole (or after mutation)
to the pGL4.20 luciferase reporter. The diagram in Fig. 3C
delineates the different constructs and knockouts (using the
Transformer kit according to Clontech’s instructions) (Fig.
3C). Certain ATGs were mutated to TTG, Inr (CATG) was
mutated to GCCG, and the TATA box of NS2 was mutated to
GCTCGAG. In addition, alternates of F1 to F7 that lacked the
luciferase initiation codon (F1 to F7) as well as two constructs
from which the NS2 core promoter was mutated (F2* and F5*)
were obtained. C6/36 and 293T (as for HeLa) cells were trans-
fected, and luciferase activity was determined. Except for F2* and
F5*, all constructs were expected to yield both NS1 and NS2
mRNAs. The observed luciferase activity in C6/36 and 293T cells
(Fig. 3E) corresponded well with the expected luciferase activities,
summarized in Fig. 3D, from (i) fusion proteins with NS1 (F1 and
F1) and NS2 (F5 and F5), (ii) directly from the luciferase
FIG 2 (A) Diagram of AalDNV expression and location of probes. (B) RNase protection assay of starts of NS and VP transcripts of AalDNV. Lanes: 1, NS probe
with a specific length of 184 nt and short nonspecific extra terminal sequences; 2, positive control for NS1 with specific size obtained with RACE experiments; 3,
positive control for NS2 with specific size obtained with RACE experiments; 4, total RNA from C6/36; 5, total RNA from AalDNV-infected C6/36 (15 g); 6, total
RNA from AalDNV-infected C6/36 (40 g); 7, VP probe; 8, positive control for VP; 9, total RNA from C6/36; 10, total RNA from AalDNV-infected C6/36 (15
g); 11, total RNA from AalDNV-infected C6/36 (40 g). The band at 101 nt confirmed the VP start, and the band at 219 nt represented protection of the VP
probe on the NS transcript. (C) Luciferase activity 40 h after transfection with different promoter constructs as fold increase over background (transfected pGL4
without insert). Inverse orientation of the promoters did not increase activity significantly over background. SV40 (not shown) and the insect virus MlDNV NS
promoter (Ml-NSP) had a strong preference for cells from vertebrates and invertebrates, respectively. Surprisingly, AalDNV had significant activity in both types
of cells.
Pham et al.
9930 jvi.asm.org Journal of Virology
on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from
initiation codon (F1 to F7), or (iii) after leaky scanning (for F1, F2,
and F5 to F7). Therefore, individual elements of both NS1 and
NS2 promoters contributed to mRNA expression.
Nevertheless, NS2 could be generated on the NS1 transcript by
leaky scanning. Blocking NS2 transcription by mutating its core
promoter in F5* yielded an 90% drop in luciferase activity in
C6/36 cells compared to F5. The remaining activity could be
ascribed to a fusion protein from ATG
NS2
on the NS1 transcript
since the introduction of an NS2 stop codon in frame after the NS2
initiation codon in F6 or knocking out ATG
NS2
in F7 abol
-
ished luciferase activity. Consequently, for F2* the remaining lu-
ciferase activity was due mainly to leaky scanning on the NS1
transcript and initiation from ATG
luc
. In conclusion, although
NS1 and NS2 mRNAs have their own promoter elements in p7/
7.4, NS1 mRNA contributes significantly through leaky scanning
to NS2 expression.
In conclusion, Northern blotting, RACE, amplicon mapping,
and RPA results were all in agreement. AalDNV used one pro-
moter region with closely overlapping elements to start transcrip-
tion of NS1 and NS2 at positions that are just 14 nt apart at either
side of ATG
NS1
. No clear TATA-like motif sequences were found
upstream of the initiator sequence CAGT of the VP of AalDNV
and AaeDNV, suggesting that these promoters were under the
control of DPE (16, 17). This regulatory circuit is likely to be one
FIG 3 Analysis of AalDNV P7/7.4 promoter elements using a luciferase reporter gene. (A) Diagram of ORFs in AalDNV, where nt 77 to 423 contain typical
sequences of core promoter elements for NS1 and NS2. (B) Three segments (I to III) were connected via pBluescript in pGL4 and could be individually mutated
and swapped. (C) In the constructs, filled boxes represent elements thought to direct NS1 mRNA expression and open boxes those thought to direct NS2 mRNA
expression, filled triangles represent transcript starts that were established, crosses represent knockouts, and gray boxes represent replaced sequences. Addition-
ally, mutants were made for all constructs in which the initiation codon of luciferase was knocked out (F1 to F7), and in 2 constructs TATA2 and Inr2 (for
NS2 transcripts) were knocked out (indicated by *). (D) Expected activities using the luciferase reporter gene in the pGL4 vector. (“l.s.” represents luciferase
activity if leaky scanning occurs.) (E) The observed luciferase activity matched the expected activity, except for the very low F7 activity (none expected). These
results indicated that the NS initiation codons, and hence the two sets of promoter elements for transcription, were individually important for expression but
nevertheless leaky scanning on the NS1 transcript could also contribute to the expression of NS2.
Expression of the AalDNV Brevidensovirus
September 2013 Volume 87 Number 17 jvi.asm.org 9931
on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from
means by which insect virus networks can transmit transcriptional
signals, such as those from DPE-specific and TATA-specific en-
hancers, via distinct pathways (18), to regulate NS and VP expres-
sion.
ACKNOWLEDGMENTS
This work was supported by a grant from the Natural Sciences and Engi-
neering Research Council of Canada to P.T. and scholarships from the
Agence Universitaire de la Francophonie and the Fondation Armand-
Frappier to H.T.P.
REFERENCES
1. Tijssen P, Agbandje-McKenna M, Almendral JM, Bergoin M, Flegel
TW, Hedman K, Kleinschmidt JA, Li Y, Pintel DJ, Tattersall P. 2011.
Parvoviridae, p 375–395. In King AMQ, Adams MJ, Carstens E, Lefkowitz
EJ (ed), Virus taxonomy: classification and nomenclature of viruses.
Ninth report of the International Committee on Taxonomy of Viruses.
Elsevier, San Diego, CA.
2. Tijssen P, Bando H, Li Y, Jousset FX, Zadori Z, Fediere G, El-Far M,
Szelei J, Bergoin M. 2006. Evolution of densoviruses, p 55– 68. In Kerr JR,
Cotmore SF, Bloom ME, Linden RM, Parrish CR (ed), Parvoviruses. Hod-
der Arnold, London, United Kingdom.
3. Bergoin M, Tijssen P. 2010. Densoviruses: a highly diverse group of
arthropod parvoviruses, p 57–90. In Asgari S, Johnson KN (ed), Insect
virology. Horizon Scientific Press, Norwich, United Kingdom.
4. Boublik Y, Jousset FX, Bergoin M. 1994. Complete nucleotide sequence
and genomic organization of the Aedes albopictus parvovirus (AaPV)
pathogenic for Aedes aegypti larvae. Virology 200:752–763.
5. Afanasiev BN, Galyov EE, Buchatsky LP, Kozlov YV. 1991. Nucleotide
sequence and genomic organization of Aedes densonucleosis virus. Virol-
ogy 185:323–336.
6. Ren X, Hoiczyk E, Rasgon JL. 2008. Viral paratransgenesis in the malaria
vector Anopheles gambiae. PLoS Pathog. 4:e1000135. doi:10.1371/journal
.ppat.1000135.
7. Jousset FX, Barreau C, Boublik Y, Cornet M. 1993. A parvo-like virus
persistently infecting a C6/36 clone of Aedes albopictus mosquito cell line
and pathogenic for Aedes aegypti larvae. Virus Res. 29:99–114.
8. Barreau C, Jousset FX, Bergoin M. 1996. Pathogenicity of the Aedes
albopictus parvovirus (AaPV), a denso-like virus, for Aedes aegypti mos-
quitoes. J. Invertebr. Pathol. 68:299–309.
9. Barreau C, Jousset FX, Bergoin M. 1997. Venereal and vertical transmis-
sion of the Aedes albopictus parvovirus in Aedes aegypti mosquitoes. Am.
J. Trop. Med. Hyg. 57:43644370.
10. Ward TW, Kimmick MW, Afanasiev BN, Carlson JO. 2001. Character-
ization of the structural gene promoter of Aedes aegypti densovirus. J.
Virol. 75:1325–1331.
11. Kimmick MW, Afanasiev BN, Beaty BJ, Carlson JO. 1998. Gene expres-
sion and regulation from the p7 promoter of Aedes densonucleosis virus.
J. Virol. 72:43644370.
12. Afanasiev BN, Kozlov YV, Carlson JO, Beaty BJ. 1994. Densovirus of
Aedes aegypti as an expression vector in mosquito cells. Exp. Parasitol.
79:322–339.
13. Tijssen P, Li Y, El-Far M, Szelei J, Letarte M, Zadori Z. 2003. Organi-
zation and expression strategy of the ambisense genome of densonucleosis
virus of Galleria mellonella. J. Virol. 77:10357–10365.
14. Liu K, Li Y, Jousset FX, Zadori Z, Szelei J, Yu Q, Pham HT, Lepine F,
Bergoin M, Tijssen P. 2011. The Acheta domesticus densovirus, isolated
from the European house cricket, has evolved an expression strategy
unique among parvoviruses. J. Virol. 85:10069–10078.
15. Fediere G, El-Far M, Li Y, Bergoin M, Tijssen P. 2004. Expression
strategy of densonucleosis virus from Mythimna loreyi. Virology 320:181–
189.
16. Kutach AK, Kadonaga JT. 2000. The downstream promoter element DPE
appears to be as widely used as the TATA box in Drosophila core promot-
ers. Mol. Cell. Biol. 20:47544764.
17. Burke TW, Willy PJ, Kutach AK, Butler JE, Kadonaga JT. 1998. The
DPE, a conserved downstream core promoter element that is functionally
analogous to the TATA box. Cold Spring Harbor Symp. Quant. Biol. 63:
75–82.
18. Hsu JY, Juven-Gershon T, Marr MT, II, Wright KJ, Tjian R, Kadonaga
JT. 2008. TBP, Mot1, and NC2 establish a regulatory circuit that controls
DPE-dependent versus TATA-dependent transcription. Genes Dev. 22:
2353–2358.
Pham et al.
9932 jvi.asm.org Journal of Virology
on June 12, 2014 by guesthttp://jvi.asm.org/Downloaded from
... Densovirus transcription has not been studied extensively. So far, VP transcripts of different densoviruses with one VP ORF are not spliced but sets of N-terminally extended proteins are generated by leaky scanning (6,(21)(22)(23). Among others, densoviruses from Blattella germanica (BgDV) and Acheta domestica (AdDV) have two VP ORFs that are joined by splicing and generate proteins with alternate N termini in addition to nested N-terminally extended sets by leaky scanning (4,24). ...
... Among others, densoviruses from Blattella germanica (BgDV) and Acheta domestica (AdDV) have two VP ORFs that are joined by splicing and generate proteins with alternate N termini in addition to nested N-terminally extended sets by leaky scanning (4,24). Leaky scanning is also important for the NS proteins of densoviruses from different genera (4,6,(21)(22)(23)(24). However, splicing determines whether NS3 or NS1/NS2 is expressed for the ambisense densoviruses, whereas the two NS genes of the brevidensoviruses have overlapping promoters so that transcripts start at either side of the NS1 start codon, allowing either NS1 or the downstream NS2 to be expressed (21)(22)(23)(24). ...
... Leaky scanning is also important for the NS proteins of densoviruses from different genera (4,6,(21)(22)(23)(24). However, splicing determines whether NS3 or NS1/NS2 is expressed for the ambisense densoviruses, whereas the two NS genes of the brevidensoviruses have overlapping promoters so that transcripts start at either side of the NS1 start codon, allowing either NS1 or the downstream NS2 to be expressed (21)(22)(23)(24). Transcription of viruses in the Iteradensovirus genus has not yet been studied. ...
... Parvoviral transcription strategies vary significantly even within each subfamily, with vertebrate-infecting PVs and ambisense DVs relying heavily on alternative splicing 3,10 . Brevihamaparvoviruses, however, display a simpler transcription profile, relying exclusively on leaky scanning 40 . AdSDV, in contrast, harbors a complex transcription strategy, which employs alternative splicing. ...
Article
Full-text available
Parvoviruses (family Parvoviridae) are currently defined by a linear monopartite ssDNA genome, T = 1 icosahedral capsids, and distinct structural (VP) and non-structural (NS) protein expression cassettes within their genome. We report the discovery of a parvovirus with a bipartite genome, Acheta domesticus segmented densovirus (AdSDV), isolated from house crickets (Acheta domesticus), in which it is pathogenic. We found that the AdSDV harbors its NS and VP cassettes on two separate genome segments. Its vp segment acquired a phospholipase A2-encoding gene, vpORF3, via inter-subfamily recombination, coding for a non-structural protein. We showed that the AdSDV evolved a highly complex transcription profile in response to its multipartite replication strategy compared to its monopartite ancestors. Our structural and molecular examinations revealed that the AdSDV packages one genome segment per particle. The cryo-EM structures of two empty- and one full-capsid population (3.3, 3.1 and 2.3 Å resolution) reveal a genome packaging mechanism, which involves an elongated C-terminal tail of the VP, “pinning” the ssDNA genome to the capsid interior at the twofold symmetry axis. This mechanism fundamentally differs from the capsid-DNA interactions previously seen in parvoviruses. This study provides new insights on the mechanism behind ssDNA genome segmentation and on the plasticity of parvovirus biology.
... The PmMDV genome, despite its 4.3-kb size, contains four predicted ORFs. Among PVs the presence of multiple, small ORFs, which frequently overlap or are encompassed by the NS or the VP cassette, are well-known (10,13,(61)(62)(63)(64)(65)(66)(67). ORF2 and ORF3 were not related to any other PV ORF, suggesting their independent acquisition by horizontal gene transfer. ...
Article
Significance Parvoviruses (PVs) are ssDNA viruses, with T = 1 icosahedral symmetry, infecting deuterostome and protostome animals. Most PVs have a highly conserved phospholipase A2 domain (PLA2) in the N-terminal region of their minor capsid protein. Under acidic pH, during endosomal/lysosomal egress, the PLA2 domain is activated to disrupt vesicle membranes. However, certain PVs lack the PLA2 and thus must use a different escape mechanism. Our study offers insight into this enigma, showing how a recently discovered PV of marine crustacean has evolved a cation-dependent mechanism to accomplish this task. We also show how host-driven convergent evolution pushed two PVs, infecting the same host species, to adopt strikingly similar surface morphologies, despite distinct multimer interactions and lack of sequence similarity.
... SDS-PAGE analysis of Bombyx mori densovirus 1 (BmDV1) show three VPs, VP1, VP2, and VP3 [153]. Penstyland Brevidensovirus transcribe only a single unspliced VP transcript resulting in a single VP that is among the smallest in the Parvoviridae at 329 aa and 358 aa, respectively [154,155]. In contrast, the Hepandensovirus cap ORF encodes a large VP1, e.g., with hepatopancreatic necrosis virus having a VP1 of 830 aa from the largest Parvoviridae genome of 6.3 kb [146,156]. ...
Article
Full-text available
Parvoviruses, infecting vertebrates and invertebrates, are a family of single-stranded DNA viruses with small, non-enveloped capsids with T = 1 icosahedral symmetry. A quarter of a century after the first parvovirus capsid structure was published, approximately 100 additional structures have been analyzed. This first structure was that of Canine Parvovirus, and it initiated the practice of structure-to-function correlation for the family. Despite high diversity in the capsid viral protein (VP) sequence, the structural topologies of all parvoviral capsids are conserved. However, surface loops inserted between the core secondary structure elements vary in conformation that enables the assembly of unique capsid surface morphologies within individual genera. These variations enable each virus to establish host niches by allowing host receptor attachment, specific tissue tropism, and antigenic diversity. This review focuses on the diversity among the parvoviruses with respect to the transcriptional strategy of the encoded VPs, the advances in capsid structure-function annotation, and therapeutic developments facilitated by the available structures.
... Interestingly, Figure 2A shows that the transcription initiation sites of NS1 and NS2 are closely positioned at either side of the NS2 initiation codon, so NS1 can be generated from the NS2 transcript by alternative initiation. This characteristic is similar to that of Aedes albopictus densovirus (AalDV) [30] during transcription. NS1 and NS2 may be encoded from separate transcripts (see Figure 3), which are transcribed by two overlapping promoters. ...
Article
Full-text available
Bombyx mori bidensovirus (BmBDV) is a single-stranded DNA (ssDNA) virus from the genus Bidensovirus of the Bidnaviridae family, which, thus far, solely infects insects. It has a unique genome that contains bipartite DNA molecules (VD1 and VD2). In this study, we explored the detailed transcription mapping of the complete BmBDV genome (VD1 and VD2) by rapid amplification of cDNA ends (RACE), reverse transcription quantitative real-time PCR (RT-qPCR), and luciferase assays. For the first time, we report the transcription map of VD2. Our mapping of the transcriptional start sites reveals that the NS genes in VD1 have separate transcripts that are derived from overlapping promoters, P5 and P5.5. Thus, our study provides a strategy for alternative promoter usage in the expression of BmBDV genes.
... Without a doubt, the DVs described so far represent only a small fraction of all the invertebrate-infecting parvoviruses that exist in nature, especially taking into account the exceptional diversity of their natural hosts. However, even the DVs that have already been described show great heterogeneity in their genome organization, size and expression strategies [1,15,45,52,59,61,65], which is paralleled by significant divergence in their genomic nucleotide sequences, the mean sequence similarity between them averaging 40 %. According to numerous recommendations on performing recombination studies, including the user's manual for the RDP4 recombination detection software utilized in this study, in order to obtain information about recombination events in a group of genomes with reasonable significance and accuracy, the genome similarity in the group should be at least 60 %, optimally ranging from 70 % to 95 % [35]. ...
Article
Full-text available
Densoviruses are a group of arthropod-infecting viruses with a small single-stranded linear DNA genome. These viruses constitute the subfamily Densovirinae of the family Parvoviridae. While recombination in between vertebrate-infecting parvoviruses has been investigated, to date, no systematic analysis of recombination has been carried out for densoviruses. The aim of the present work was to study possible recombination events in the evolutionary history of densoviruses and to assess possible effects of recombination on phylogenies inferred using amino acid sequences of nonstructural (NS) and capsid (viral protein, VP) proteins. For this purpose, the complete or nearly complete genome nucleotide sequences of 40 densoviruses from the GenBank database were used to construct a phylogenetic cladogram. The viruses under study clustered into five distinct groups corresponding to the five currently accepted genera. Recombination within each group was studied independently. The RDP4 software revealed three statistically highly credible recombination events, two of which involved viruses of the genus Ambidensovirus, and the other, viruses from the genus Iteradensovirus. These recombination events led to mismatches between phylogenetic trees constructed using comparison of amino acid sequences of proteins encoded by genome regions of recombinant and non-recombinant origin (regulatory NS1 and NS3 proteins and capsid VP protein).
Article
A wide spectrum of invertebrates is susceptible to various single-stranded DNA viruses. Their relative simplicity of replication and dependence on actively dividing cells makes them highly pathogenic for many invertebrates (Hexapoda, Decapoda, etc.). We present their taxonomical classification and describe the evolutionary relationships between various groups of invertebrate-infecting viruses, their high degree of recombination, and their relationship to viruses infecting mammals or other vertebrates. They share characteristics of the viruses within the various families, including structure of the virus particle, genome properties, and gene expression strategy.
Article
Herein, we report the identification of putative promoters for the non-structural proteins (NS) and capsid structural proteins (VP) of Helicoverpa armigera densovirus (HaDV2) as well as a potential mechanism for how these promoters might be regulated. For the first time, we report that VP is able to transactivate the VP promoter and, to a lesser degree, the NS promoter in densoviruses. In addition to this, another promoter-like sequence designated P2, when co-transfected with the VP gene, enhanced luciferase activity by approximately 35 times compared to a control. This suggests that there are two promoters for VP in HaDV2 and that the VP of parvoviruses might play a more important role in viral transcription than previously appreciated.
Article
A wide spectrum of invertebrates is susceptible to various single-stranded DNA viruses. Their relative simplicity of replication and dependence on actively dividing cells makes them highly pathogenic for many invertebrates (Hexapoda, Decapoda, etc.). We present their taxonomical classification and describe the evolutionary relationships between various groups of invertebrate-infecting viruses, their high degree of recombination, and their relationship to viruses infecting mammals or other vertebrates. They share characteristics of the viruses within the various families, including structure of the virus particle, genome properties, and gene expression strategy.
Chapter
The transmission of mosquito-vectored human pathogens has continued to exert a global public health burden despite control measures. Recent advances in our understanding of the mosquito microflora have demonstrated that microbes play a major role in determining the outcome of mosquito infection by human pathogens. Characterization of mosquito symbionts has elucidated molecular mechanisms responsible for the inhibition of pathogen infection and has provided natural microbial candidates for the development of strategies to interrupt disease transmission. Furthermore, some mosquito symbionts can also be genetically modified to enhance their ability to antagonize pathogen infection in a process known as paratransgenesis, a next-generation approach to interrupt the transmission of mosquito-borne disease.
Article
Full-text available
The Acheta domesticus densovirus (AdDNV), isolated from crickets, has been endemic in Europe for at least 35 years. Severe epizootics have also been observed in American commercial rearings since 2009 and 2010. The AdDNV genome was cloned and sequenced for this study. The transcription map showed that splicing occurred in both the nonstructural (NS) and capsid protein (VP) multicistronic RNAs. The splicing pattern of NS mRNA predicted 3 nonstructural proteins (NS1 [576 codons], NS2 [286 codons], and NS3 [213 codons]). The VP gene cassette contained two VP open reading frames (ORFs), of 597 (ORF-A) and 268 (ORF-B) codons. The VP2 sequence was shown by N-terminal Edman degradation and mass spectrometry to correspond with ORF-A. Mass spectrometry, sequencing, and Western blotting of baculovirus-expressed VPs versus native structural proteins demonstrated that the VP1 structural protein was generated by joining ORF-A and -B via splicing (splice II), eliminating the N terminus of VP2. This splice resulted in a nested set of VP1 (816 codons), VP3 (467 codons), and VP4 (429 codons) structural proteins. In contrast, the two splices within ORF-B (Ia and Ib) removed the donor site of intron II and resulted in VP2, VP3, and VP4 expression. ORF-B may also code for several nonstructural proteins, of 268, 233, and 158 codons. The small ORF-B contains the coding sequence for a phospholipase A2 motif found in VP1, which was shown previously to be critical for cellular uptake of the virus. These splicing features are unique among parvoviruses and define a new genus of ambisense densoviruses.
Article
Full-text available
Paratransgenesis, the genetic manipulation of insect symbiotic microorganisms, is being considered as a potential method to control vector-borne diseases such as malaria. The feasibility of paratransgenic malaria control has been hampered by the lack of candidate symbiotic microorganisms for the major vector Anopheles gambiae. In other systems, densonucleosis viruses (DNVs) are attractive agents for viral paratransgenesis because they infect important vector insects, can be genetically manipulated and are transmitted to subsequent generations. However, An. gambiae has been shown to be refractory to DNV dissemination. We discovered, cloned and characterized the first known DNV (AgDNV) capable of infection and dissemination in An. gambiae. We developed a flexible AgDNV-based expression vector to express any gene of interest in An. gambiae using a two-plasmid helper-transducer system. To demonstrate proof-of-concept of the viral paratransgenesis strategy, we used this system to transduce expression of an exogenous gene (enhanced green fluorescent protein; EGFP) in An. gambiae mosquitoes. Wild-type and EGFP-transducing AgDNV virions were highly infectious to An. gambiae larvae, disseminated to and expressed EGFP in epidemiologically relevant adult tissues such as midgut, fat body and ovaries and were transmitted to subsequent mosquito generations. These proof-of-principle data suggest that AgDNV could be used as part of a paratransgenic malaria control strategy by transduction of anti-Plasmodium peptides or insect-specific toxins in Anopheles mosquitoes. AgDNV will also be extremely valuable as an effective and easy-to-use laboratory tool for transient gene expression or RNAi in An. gambiae.
Article
The RNA polymerase II core promoter is a structurally and functionally diverse transcriptional module. RNAi depletion and overexpression experiments revealed a genetic circuit that controls the balance of transcription from two core promoter motifs, the TATA box and the downstream core promoter element (DPE). In this circuit, TBP activates TATA-dependent transcription and represses DPE-dependent transcription, whereas Mot1 and NC2 block TBP function and thus repress TATA-dependent transcription and activate DPE-dependent transcription. This regulatory circuit is likely to be one means by which biological networks can transmit transcriptional signals, such as those from DPE-specific and TATA-specific enhancers, via distinct pathways.
Article
Over 99% of the genome of Aedes densonucleosis virus was determined. Two types of the viral DNA were found that differ only in four nucleotides (nt) in the 5' noncoding part and whose sizes are 4009 nt (more copious) and 4012 nt, respectively. Both 146 nt at the 3' end and 164 nt at the 5' end could assume a similar T-shaped structure; but unlike the adeno-associated virus, Aedes DNA has a unique primary DNA sequence at each terminus. However, the crossarms of these structures are built of the same sequences. An imperfect direct repeat of 34 nt was observed in the 5' noncoding part. The plus strand has three large open reading frames (ORF): a left ORF, a right ORF, and a mid ORF (within the left ORF). The left ORF codes for the nonstructural protein NS-1 (97.5K) featured by an NTP-binding domain, and the right ORF encodes the both capsid proteins, the smaller of which (39K) is supposed to be derived from the larger one (40.5K) by proteolytic cleavage. There is also an ORF in the minus strand. The putative polypeptide coded by this ORF is extremely hydrophobic.
Article
We have constructed an infectious DNA clone containing the genome of Aedes aegypti densovirus (AeDNV) in a bacterial plasmid. When this clone was transfected into Aedes albopictus C6/36 mosquito cells, the AeDNV genome rescued from the plasmid and replicated as the wild-type virus. To investigate the cloned virus as an expression vector, the reporter gene encoding beta-galactosidase (beta-gal) was inserted into four large open reading frames (ORF) observed in the AeDNV genome. When these recombinant constructs were transfected into Aedes albopictus C6/36 cells, the beta-gal was expressed efficiently from the right ORF (encoding capsid proteins, Vps) and the mid ORF (encoding putative nonstructural protein 2). A low level of expression was found from the left ORF (encoding nonstructural protein 1, NS1), and no expression was detected from the ORF observed on the minus strand of the AeDNV genome. The expression from the right, mid, and left ORFs can be trans-activated with NS1. A putative nuclear targeting sequence observed in the N-terminus of the AeDNV Vps is presumed to be responsible for transport of the chimeric beta-gal into nucleus. The recombinant AeDNV genomes (carrying the beta-gal gene) supplied with the AeDNV capsid proteins can be packaged into infectious transducing particles. Our results indicate that the genome of AeDNV can serve as a vector for delivery and expression of foreign genes in mosquito cells with subsequent targeting of the product to the desired cell compartment.
Article
We have cloned the replicative form of the Aedes albopictus parvovirus (AaPV) genome and determined the complete sequence of the viral strand. The sequence is 4176 nucleotides (nt) in length. The first 134 nt at the 3' end and the terminal 182 nt at the 5' end of the viral (minus) strand can both generate by folding and annealing of complementary sequences a typical terminal T-shaped structure although they differ in their sequence. Three large open reading frames (ORFs), each one in a different frame, are present between map units (mu) 8.0 and 87.6 on the complementary (plus) strand. The left, mid (located within the left ORF), and right ORFs have potential coding capacities of 95, 41, and 40 kDa, respectively. Two potential promoters were found upstream from the left and right ORFs, at mu 7.2 and mu 60.0, respectively. Computer search for sequence homologies suggests that the left ORF very likely encodes the nonstructural NS-1 protein since it contains the highly conserved NTP-binding amino acid (aa) domain (GKRN sequence) of all parvoviruses. Comparison with other invertebrate and vertebrate parvoviruses revealed that the AaPV genome shares 77.3% nt sequence homology and between 73 and 78% aa sequence homologies with the Aedes aegypti densonucleosis virus (Aedes DNV). Organization of both genomes was similar except that no potential ORF was found on the minus strand of AaPV. The difference of 167 nt in length between AaPV and Aedes DNV (4009 nt) genomes is due to additional noncoding sequences located between the internal coding region and the terminal palindromes in the AaPV genome. No significant homology was found between AaPV and the two other insect parvoviruses sequenced so far, the Bombyx mori DNV (BmDNV) and the Junonia coenia DNV (JcDNV).