Tài liệu Báo cáo khoa học: Nuclear receptors in the mosquito Aedes aegypti Annotation, hormonal regulation and expression profiling - Pdf 10

Nuclear receptors in the mosquito Aedes aegypti
Annotation, hormonal regulation and expression profiling
Josefa Cruz*, Douglas H. Sieglaff*,, Peter Arensburger, Peter W. Atkinson and Alexander S. Raikhel
Department of Entomology and the Institute for Integrative Genome Biology, University of California, Riverside, CA, USA
Mosquitoes are vectors of some of the world’s most
devastating diseases. Malaria causes approximately
1 million deaths annually (http://www.who.org) and
dengue, a rapidly expanding disease in most tropical
and subtropical areas of the world, has become the
most significant arboviral disease of humans. Anophe-
line mosquitoes are the vectors of malaria, whereas
Aedes species is the vector of dengue and yellow fever.
Both disease vectors are exquisitely adapted to living
around humans and using human blood as a nutrient
source to promote egg development. A basic under-
standing of mosquito reproductive biology is an
important component in developing novel strategies
for use in the control of mosquito-borne disease.
Egg maturation in Aedes aegypti adult females
includes a process termed vitellogenesis, which involves
massive production of yolk protein precursors (YPPs)
by the fat body and their subsequent internalization
into the developing oocyte, helping to support later
embryonic development. The two primary insect
Keywords
ecdysone receptor; 20-hydroxyecdysone;
nuclear receptor; reproduction; yolk protein
Correspondence
A. S. Raikhel, Department of Entomology,
University of California, Riverside, Watkins
Drive, CA 92521, USA

conducted to identify the role of the steroid hormone 20-hydroxyecdysone
in modulating the expression of A. aegypti NRs. These data which describe
the identification, expression and hormonal regulation of 20 NRs in the
yellow fever mosquito lay a solid foundation for future studies on the
hormonal regulation of reproduction in mosquitoes.
Abbreviations
Chx, cycloheximide; 20E, 20-hydroxyecdysone; EcR, ecdysone receptor; JH III, juvenile hormone III; NR, nuclear receptors; PBM, post blood
meal; Vg, vitellogenin; YPP, yolk protein precursors.
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1233
hormones governing vitellogenesis are the sesquiterpe-
noid juvenile hormone III (JH III) and the steroid 20-
hydroxyecdysone (20E). The period following adult
eclosion requires JH III to promote the development
of ‘competence’ or the ability of the female mosquito
to process the blood meal in the promotion of vitello-
genesis [1,2]; 72 h is typically required after eclosion to
achieve this state. This development toward ‘compe-
tence’ is termed pre-vitellogenesis. JH III titer levels
are highest in adult females during pre-vitellogenic
development, but fall dramatically after ingestion of
the blood meal; the 20E concentration, however,
begins to increase within a few hours post blood meal
(PBM), peaking at 18–24 h PBM [3]. 20E is one of the
primary regulators in the synthesis of vitellogenin (Vg),
the main YPP protein produced by the fat body [4,5].
The molecular mechanism of 20E action has been dis-
sected in detail in studies of Drosophila melanogaster
development [6–9]. The functional 20E receptor is com-
posed of two proteins, the ecdysone receptor (EcR),
which binds specifically to 20E, and the product of the

p160 ⁄ SRC (AaFISC) which, in turn, binds the
AaEcR ⁄ AaUSP heterodimers, establishing a functional
multiple protein complex on the Vg promoter [16]. By
24 h PBM, AaVg transcript levels reach their maxi-
mum, after which they sharply decline, concluding
with the termination of vitellogenesis. In this termina-
tion process, mosquito Seven-up (AaSvp), a NR mem-
ber, plays a central role replacing AaUSP in the
AaEcR ⁄ AaUSP heterodimer complex, thereby block-
ing the action of 20E [13]. Another A. aegypti NR,
AaHNF-4c, has also been proposed to promote the
termination of Vg expression [17], but the mechanism
is still unknown. The regulation of AaVg gene
expression by 20E acts not only through members of
the NR family, but also through other transcription
factors such as E74, Ets-domain protein and Broad-
complex, C2H2-type zinc-finger DNA-binding protein
[18,19].
Despite the achievements mentioned so far, there are
additional NRs that remain uncharacterized in the
mosquito, some of which may play an important role
in its reproduction. In this study, we identified and
began to characterize all putative NRs of A. aegypti.
We report the annotation of 19 canonical NR family
members along with one member of the so-called
Knirps group. In addition, we determined the expres-
sion profiles for transcripts of these NRs within two
reproductive tissues of the adult female mosquito – the
fat body and ovaries. Furthermore, using an in vitro
fat body culture system allowed us to identify NRs

The competence factor bFTZ-F1 of A. aegypti was
first cloned by Li et al. [23] from a cDNA library
prepared from the fat bodies of vitellogenic female
mosquitoes. They isolated several clones that code
for a single protein. Interestingly, during the initial
process of identifying the NR family members in the
A. aegypti genome, we predicted two isoforms of
AaFTZ-F1, differing only in their A ⁄ B region, as
observed for the D. melanogaster FTZ-F1 isoforms
[24,25]. Initially, we hypothesized that the mosquito
isoforms would be related to those described for
D. melanogaster, but with a low percentage of identity
in the A ⁄ B domain between the two (data not shown).
We designated the isoforms of this A. aegypti NR as
AabFTZ-F1A (previously named AabFTZ-F1) [23]
and AabFTZ-F1B.
Phylogenetic analysis
We conducted a phylogenetic analysis of the NRs from
the five insect genomes sequenced so far: D. melano-
gaster, Anopheles gambiae, Ap. mellifera, T. castaneum
and A. aegypti as well as the sequences of the human
orthologs. When different isoforms were recovered,
only the longest amino acid sequences that included
the DNA binding, hinge and ligand-binding domains
were used for this analysis, except for the Knirps fam-
ily members that lack the LBD. In D. melanogaster,
Table 1. NRs of Aedes aegypti. NR family members according to the NuReBASE proposed nomenclature [27]. General names are based on
the nomenclature of D. melanogaster with the exception of PNR-like, which has not been annotated in D. melanogaster, and is named
according to the Ap. mellifera ortholog. A. aegypti NR name and isoform, if present. VectorBase and NCBI Accession numbers. The NRs
transcriptionally controlled by 20E, in an in vitro fat body culture system are indicated in the 20E response column. NT, not tested; P, primary

AF305214 S
NR2D1 Hormone receptor-like in 78 AaHr78 AAEL001796 BN001173 P
NR2E2 Tailless AaTll AAEL003020 BN001174 NR
NR2E3 Hormone receptor-like in 51 AaHr51 AAEL007190 BN001175 NT
NR2E4 Dissatisfaction AaDsf AAEL005381 BN001178 NT
NR2E5 Hormone receptor-like in 83 AaHr83 AAEL007350 AM773444 NT
NR2E6 PNR-like AaPNR-like AAEL008043, AAEL008047 AM773445 NR
NR2F3 Seven up AaSvp AAEL006916, AAEL002765,
AAEL002768
AF303224 NR
NR3B4 Estrogen-related receptor AaERR AAEL013546 BN001176 NR
NR4A4 Hormone receptor-like in 38 AaHr38 AAEL013270 AF165528 NR
NR5A3 Ftz transcription factor AaFTZ-F1 AabFTZ-F1A AAEL002053, AAEL002062 AF274870 NR
AabFTZ-F1B AAEL002062 AM773446 S
NR5B1 Hormone receptor-like in 39 AaHr39 AAEL001304 BN001177 P
NR6A1 Hormone receptor-like in 4 AaHr4 AAEL005850, AAEL005864 AM773447 P
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1235
this group is composed of three members (knirps,
knirps-like and eagle). In A. aegypti, we only charac-
terized one member of the Knirps group that presented
higher amino acid similarity with Dmknirps-like (31%)
than with Dmknirps (23%) or DmEg (25%, data not
shown). Remarkably, both A. aegypti and An. gambiae
genomes contain only one Knirps family member
(knirps-like, Table S1).
As previously mentioned, we identified 19 canoni-
cal NR family members in the A. aegypti genome, 18
of which are likely orthologs of D. melanogaster, An.
gambiae and Ap. mellifera NRs, and one which is a

reported [30,31].
Expression in adult female reproductive tissues
To determine whether NR family members are
expressed in the two main reproductive tissues of adult
female mosquitoes, we conducted an initial assessment
using RT-PCR. Total RNA was extracted from the fat
body and ovaries of pre-vitellogenic females 5–6 days
after eclosion and from vitellogenic females 6–12 and
18–24 h after a blood meal, and then was subjected to
RT-PCR with a specific primer pair for each NR (see
Table S2 for primer sequences). Two biological repli-
cates were analyzed, and Fig. 2 depicts the profile
matching both replicates.
An increase in transcript abundance for AaEcRA,
AaEcRB, AaE75A, AaE75B, AaE75C, AaHR3,
AaHR4, AaE78 and AaHR39 occurred in both tissues,
correlating with the known rise in ecdysteroids in vivo ,
whereas AaUSP-B, AaHR38, AaTll and AaPNR-like
only displayed an increase in transcript abundance in
the fat body during this same period (Fig. 2). This sug-
gests that these later orphan NRs may be hormone
inducible, which we addressed using in vitro assays
(see below). Many NR transcripts displayed a decrease
during the initial phase of vitellogenesis (6 + 12 h
PBM) only to increase again at peak vitellogenesis
(18 + 24 h PBM). These transcripts (AaUSP-A,
AabFTZ-F1A
, AabFTZ-F1B, AaHR78, AaHNF-4A,
AaHNF-4B, AaHNF-4C, AaSvp and AaERR) were
also analyzed in our in vitro assay. Any kind of fluctu-

As observed in Fig. 2, the majority of NRs are
expressed at a constant level within the ovaries during
the period analyzed. However, AaE75A, AaE75B,
Nuclear receptors in Aedes aegypti J. Cruz et al.
1236 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
Fig. 1. Phylogenetic tree of insect NRs. The different NR families are organized into groupings, NR1–NR6. The tree was constructed follow-
ing the distance-based neighbor-joining method, using the NRs sequences of D. melanogaster (Dm), A. aegypti (Aa), An. gambiae (Ag),
Ap. mellifera (Am) and T. castaneum (Tc) indicated in Table S1 as well as the human (H. sapiens, Hs) orthologs obtained from Genebank.
Branch lengths are proportional to sequence divergence. The bar represents 0.1 substitutions per site. The bootstraps nodal support values
are shown.
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1237
AaE75C and AaHR3 mRNAs increased with the
in vivo ecdysteroid peak, corroborating results from
previous studies [36,37]. Such an increase in expres-
sion levels within the ovary along with known ecdys-
teroid titers in vivo was also observed for AaEcRA,
AaEcRB, AaHR4, AaE78, AaHNF-4A and AaHNF-
4C transcripts. AaSvp was the only one that displayed
a reduction in mRNA levels in ovaries at 18–24 h
PBM (Fig. 2). While the ecdysone response hierarchy
has been extensively studied in development and
metamorphosis in insects, its potential role in promot-
ing oogenesis has received significantly less attention.
EcR mutant females of D. melanogaster display
abnormal egg chamber development and loss of vitell-
ogenic egg stages [38] as well as chorion malforma-
tions [39]. In the cockroach Blattella germanica,
females treated with BgEcRA dsRNA displayed a
reduction in the number of follicular cells in the basal

against those NRs that had displayed dynamic expres-
sion within the fat body in our earlier RT-PCR experi-
ment. Total RNA was isolated from the fat body of
three independent collections of mosquito females
staged at different time points during pre-vitellogenic
and vitellogenic stages. The same amount of RNA was
retro-transcribed and analyzed by means of qPCR
using specific primer pairs for each NR (Table S2).
In vivo fat body NR transcript expression levels were
standardized by total RNA input, because the fat body
is a dynamically developing tissue both before and
following a blood meal, thus precluding the use of a
Fig. 2. Expression of A. aegypti nuclear receptors (NRs) in fat body
(FB) and ovary (Ov) of pre-vitellogenic female mosquitoes (PV)
4–5 days after eclosion and at 6–12 or 18–24 h after a blood meal
(PBM) was determined by quantitative PCR. The profiles are repre-
sentative of two biological replicates.
Nuclear receptors in Aedes aegypti J. Cruz et al.
1238 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
AaHR3
0
500
1000
1500
2000
2500
b
ab
a
a

a
a
a
a
0
100
200
AaEcR-A
0
20
40
60
80
100
120
b
a
b
b
b
b
b
b
0 h 6 h 12 h 24 h 36 h 48 h 72 h
20E (pg/female)
50
100
150
200
250

a
a
a
a
a
AaE75A
1 d 4 d 6 h 12 h 24 h 36 h 48 h 72 h
0
400
800
1200
1600
b
a
b
b
b
b
ab
b
PV PBM
1 d 4 d 6 h 12 h 24 h 36 h 48 h 72 h
PV PBM
PV
AB
PBM
AaEcR-B
0
1
2

are expressed as relative mRNA and are the mean of three independent biological replicates. The vertical bars indicate the SEM. Means
were separated using Tukey–Kramer HSD with time points sharing the same letter determined not to be significantly different (P £ 0.05).
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1239
‘normalizing’ transcript. The time points chosen for
the current study address the complete vitellogenic
cycle: pre-vitellogenesis (1–4 days pre-vitellogenesis),
vitellogenesis (6–30 h PBM), early post vitellogenesis
(36–48 h PBM) and late post vitellogenesis (72 h
PBM), with these progressions including, respectively,
active ribosomal biogenesis, massive protein synthesis,
tissue autophagy and ribosomal biogenesis again
[2,45,46]. This developmental course can be observed
through the dynamic expression profile of the com-
monly used ‘housekeeping’ transcript ribosomal pro-
tein S7 [47] (Fig. 3E), as well as actin (Fig. 3F). Such a
condition is not applicable to the in vitro experiments,
because all fat bodies used in these studies were at the
AaERR
10
20
30
40
50
a
a
a
a
a
a

10
a
a
a
a
a
a
a
a
10
AaHNF4B
0
2000
4000
6000
8000
10 000
b
a
b
b
b
b
ab
a
AaHNF4C
0
500
1000
1500

100
150
200
250
50
100
150
200
250
0 d 4 d
PV
CD
PBM
0 h 6 h 12 h 24 h 36 h 48 h 72 h
20E (pg/female)
0 d 4 d
PV PBM
Aa
β
FTZ-F1A
20
40
60
80
100
120
a
b
ab
b

to determine whether the steroid hormone 20E might
be responsible for the expression profile observed
in vivo. To this end, we carried out two different
in vitro experiments. First, our aim was to establish
those NRs directly induced by 20E (primary-response
genes). The fat bodies were incubated in the presence
of 20E alone, cycloheximide (Chx) alone, 20E plus Chx
or control media for 6 h. In the second experiment, our
aim was to determine the NR transcripts that require
an initial exposure to 20E followed by its withdrawal
for induction (secondary-response genes). In this sec-
ond experiment, the fat bodies were incubated in media
supplemented with 20E for 4 h, washed and then incu-
bated for 12 more hours in a hormone-free medium.
As a control, fat bodies were incubated with or without
20E. RNA extracted from these samples was analyzed
by means of qPCR, and transcripts were normalized
over AaS7. A summary of the 20E inducibility of the
transcripts analyzed is presented in Table 1.
The A. aegypti ecdysone receptor
Two EcR isoforms (AaEcRA and AaEcRB) and two
USP isoforms (AaUSP-A and AaUSP-B) have been
characterized previously in A. aegypti [48–50]. The
Relative mRNA (+SEM)
Relative mRNA (+SEM)
AaActin
0
50
100
150

40
60
80
100
120
140
a
a
a
a
a
a
a
a
10
20
AaSvp
0
20
40
60
80
a
a
a
a
a
a
a
a

b
b
b
b
b
PV
EF
PBM
0 h 6 h 12 h 24 h 36 h 48 h 72 h
20E (pg/female)
50
100
150
200
250
20E (pg/female)
50
100
150
200
250
0 d 4 d0 h 6 h 12 h 24 h 36 h 48 h 72 h0 d 4 d
PV PBM
Fig. 3. (Continued).
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1241
expression pattern of AaEcRA followed the peak in
20E titers, as previously reported [48]. The level of
AaEcRA transcript increased slightly at 12 h PBM,
reaching the maximum at 24 h (Fig. 3A), an 8 h delay

PBM [49] was not observed. The level of AaUSP-B
transcript was relatively constant throughout the pre-
and vitellogenic periods, with a slight increase begin-
ning at the end of the vitellogenic period (48–72 h;
Fig. 3A). Previous 20E transcriptional regulation ana-
lysis in this laboratory, using semi-quantitative RT-
PCR, showed that AaUSP-A mRNA was upregulated
after a short exposure to 20E and its withdrawal [49],
a response that we could not corroborate using qPCR
(Fig. 4A). Furthermore, AaUSP-A mRNA levels only
increased significantly when incubated with Chx in the
absence of 20E (Fig. 4A). By contrast, AaUSP-B tran-
scripts were upregulated by 20E in combination with
Chx, but also after a long exposure to 20E alone [49].
Our current results, along with previous reports that
have established a lack of fluctuation in the protein
levels of AaEcR and AaUSP isoforms during vitello-
genesis [13], suggest a lack of significant transcriptional
and translational regulation of the two components
that make up the ecdysone receptor. Moreover,
co-immunoprecipitation experiments using nuclear
extracts of vitellogenic fat bodies of A. aegypti demon-
strated that the formation of the heterodimer AaE-
cR ⁄ AaUSP can be regulated by other NRs through
protein–protein interactions. AaHR38 and AaSvp,
during the arrest and termination of vitellogenesis,
respectively, bind to AaUSP preventing its hetero-
dimerization with AaEcR, and, consequently, the 20E-
dependent activation is blocked. The presence of 20E,
however, favors the formation of the AaEcR⁄ AaUSP

The primary difference between the observed tran-
script profiles of AaHR3 and AaHR4 is that AaHR4 is
significantly abundant in newly eclosed female fat
bodies (pre-vitellogenic 1 day, Fig. 3B), suggesting a
possible role in the transition between pupae and
adult. It is generally believed that gene transcripts
induced in vitro only in the presence of 20E and
the protein synthesis inhibitor Chx, as observed for
AaHR4 and AaHR3, are negatively repressed by
20-induced genes (e.g. the early response genes) [53,54].
M. sexta GV1 cell transfection assays demonstrated
Nuclear receptors in Aedes aegypti J. Cruz et al.
1242 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
Relative mRNA (SEM)
Relative mRNA (SEM)
0
2
4
6
8
10
12
14
16
18
**
AaUSPA
0 h
CMCM 20E Chx
20E + Chx

AaEcRB
0
200
400
600
800
1000
1200
1400
1600
1800
AaUSPA
0
2
4
6
8
10
12
14
16
18
25
20
10
15
0
5
AaUSPB
**

**
5
4
2
3
0
1
6
AaE75A
**
****
Fig. 4. Effect of 20E on the transcription of A. aegypti nuclear receptors (NRs) in isolated pre-vitellogenic (PV) fat bodies. In the first experi-
ment (left panel), PV fat bodies dissected from female mosquitoes (5 days) were incubated in culture media (CM) for 30 min and established
as initial time point (0 h); then were incubated for 6 h in CM, with 10
)6
M 20E (20E), 10
)5
M Chx (Chx) or 20E and Chx together
(20E + Chx). In the second experiment (right), 5 day PV fat bodies were incubated in CM for 30 min and established as initial time point
(0 h); then were incubated in CM or with 20E for 4 and 16 h, or with a pulse treatment of 20E for 4 h then removal of 20E, followed by incu-
bation within CM for an additional 12 h (4 h 20E + CM). At the indicated time points, a group of nine fat bodies were collected and RNA lev-
els were analyzed using qPCR. Transcript abundance values for AaEcRA, AaEcRB, AaUSP-A, AaUSP-B, AaE75A (A), AaHR3, AaHR4, AaE78,
AaHR39, AaHR78 (B) and AabFTZ-F1A, AabFTZ-F1B, AaHNF-4A, AaHNF-4B, AaHNF-4C (C) are presented as mRNA quantity normalized
against ribosomal protein S7 transcripts and represent the mean of three independent biological replicates. The vertical bars indicate the
SEM. Each normalized transcript was compared against the 0 h CM using Dunnett’s method. Asterisks indicate statistically significant differ-
ences at ****P £ 0.0005; ***P £ 0.005; **P £ 0.05; *P £ 0.1 levels.
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1243
that MsE75A represses the induction of MsHR3 by
binding to the consensus monomeric response element

6
8
10
0
1
2
3
4
5
6
AaHR78
**
AaHR78
0
1
2
3
4
5
6
0
2
4
6
8
10
AaHR39
****
*
0

8
10
12
14
AaHR4
***
0
2
4
6
8
AaHR3
B
****
AaHR3
0
2
4
6
8
**
**
Fig. 4. (Continued).
Nuclear receptors in Aedes aegypti J. Cruz et al.
1244 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
importantly, are involved in coordinating the cascade
of expression of various NRs. In D. melanogaster,
DmE75B represses DmHR3-dependent transactivation
through protein–protein interaction [59]. DmHR3 and
DmHR4 act together to induce DmbFTZ-F1 expres-

****
***
Aa
β
FTZ-F1A
C
0
10
20
30
40
50
**
AaHNF4A
0
40
80
120
160
200
AaHNF4C
Aa
β
FTZ-F1A
0
20
40
130
140
**

120
160
200
0
100
200
300
400
Aa
β
FTZ-F1B
**
*
0
2
4
6
8
10
12
AaHNF4B
*
Fig. 4. (Continued).
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1245
the ecdysteroid-induced transcription factors. Such cas-
cades will likely differ among species, or even physio-
logical processes. In A. aegypti, the discrete expression
of both AaHR3 and AaHR4 suggests their function in
the regulatory hierarchy in the mosquito fat body dur-

Transcript levels of AaHR39 were highest after eclo-
sion and in post-vitellogenic fat bodies, and lowest
during vitellogenesis (Fig. 3C). 20E was capable of
inducing the transcription of AaHR39, with a much
more significant increase in the presence of Chx
(Fig. 4B). This finding agrees with that observed
in vivo, in which transcript abundance increased later
in the vitellogenic cycle. HR39, a NR with high
sequence similarity to FTZ-F1, has been identified in
other insect species, D. melanogaster, B. mori and
T. castaneum [21,67–69]. In D. melanogaster, both
DmbFTZ-F1 and DmHR39 mRNAs are expressed dur-
ing the same developmental stages; however, the
expression of DmHR39 typically precedes DmbFTZ-F1
and seems to be downregulated when DmbFTZ-F1
reaches its maximum levels [7,70]. Both have similar
DNA-binding domains, and DmHR39 represses the
transcription activated by DmbFTZ-F1 through bind-
ing to the same response element [67,71]. Moreover,
a GAL4-LBD ‘ligand sensor’ system showed that
DmHR39 does not display detectable activation at any
stage of development, suggesting that this NR acts as
a repressor [72]. It would be interesting to determine
whether the reciprocal patterns of expression between
AaHR39 and both AabFTZ-F1 isoforms during the
vitellogenic period are of functional significance.
The period during which AaHR78 displayed its
maximum levels correlates with the pre-vitellogenic
preparatory period in the fat body, being low with
higher titers of 20E and gradually rising again after

A ⁄ B-specific region of AabFTZ-F1A [15]. As a result,
the experiments conducted using this antibody are
specific for AabFTZ-F1A isoform. For the current
study, we determined whether the two isoforms dis-
played different expression profiles and are both regu-
lated by 20E in a similar manner. As shown in
Fig. 3C, the level of AabFTZ-F1A transcript abun-
dance was significantly higher than that of AabFTZ-
F1B in the fat bodies of newly eclosed females. After
Nuclear receptors in Aedes aegypti J. Cruz et al.
1246 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
the onset of vitellogenesis, AabFTZ-F1A dropped dra-
matically, remaining at low levels until vitellogenesis
was complete (36–72 h PBM). AabFTZ-F1B mRNA
displayed near background levels of transcript abun-
dance during pre-vitellogenic development and
throughout vitellogenesis, with expression levels only
beginning to rise following vitellogenesis (36 h PBM;
Fig. 3C). Experiments with in vitro fat body cultures
revealed that AabFTZ-F1A expression is not directly
under 20E regulation (Fig. 4C), and its transcript lev-
els only reached significant quantities when exposed to
the protein synthesis inhibitor Chx. This observation
could be explained by a stabilization of pre-existing
mRNAs [53,54,77] or due to Chx inhibiting the
expression of repressor factors. In D. melanogaster,
the NR DmbFTZ-F1 has been defined as a ‘compe-
tence’ factor due to the requirement for its expression
during mid-prepupal development, allowing for the
correct response to ecdysone at the end of the pupal

agreement with studies of D. melanogaster [78], B. mori
[82] and M. sexta [56]. In D. melanogaster, DmHR3
activates DmbFTZ-F1 mRNA expression through
a response element in the promoter region of the
DmbFTZ-F1
gene [59,60,83]. But DmE75B, which
lacks a complete DNA-binding domain, inhibits this
inductive function by forming a complex with DmHR3
on the DmbFTZ-F1 promoter. This mechanism pro-
vides specific timing for DmbFTZ-F1 transcription that
requires the presence of 20E for DmHR3 induction,
but its withdrawal for the disappearance of DmE75B
[59,60]. In vivo, maximum expression of AabFTZ-F1B
occurred after peak AaE75 and AaHR3 expression,
suggesting a similar regulatory activation cascade in
the vitellogenic mosquito fat body (Fig. 3B,C). Further
studies are necessary to clarify the regulation of
AabFTZ-F1B and its possible involvement in the
cascade as ecdysteroid titers decline later in the vitello-
genic cycle.
Expression and 20E regulation of A. aegypti
HNF-4 isoforms
There are three isoforms of the HNF-4 in A. aegypti,
which have been previously designated AaHNF-4A,
AaHNF-4B and AaHNF-4C [17]. The A and B iso-
forms are typical members of the NR family, differing
only in the N-terminal end of the variable A ⁄ B
domain. The third mosquito isoform, AaHNF-4C,
lacks the greater part of this A ⁄ B domain and the
complete DBD; consequently, it cannot bind DNA

enzymes that are activated at fasting and suppressed in
a fed state [88]. In D. melanogaster, a recent study
using an in vivo ligand-detection system that follows
NR LBD activation patterns in vivo, by way of a
GAL4-LBD system [89], showed that the activity
of GAL4-DmHNF-4, along with GAL4-DmHR3 and
GAL4-DmHR38, in fat body is dramatically downreg-
ulated at puparium formation. This coincides with the
cessation of feeding that occurs at the end of the larval
development [72]. Thus, it was concluded that these
NRs might respond to nutrients or metabolites. In
A. aegypti, all three AaHNF-4 isoforms are upregula-
ted at the end of the vitellogenic period, a time when
the female has fully digested and processed the blood
meal, and once again begins a fasting period until the
next blood feed. By contrast to what is observed
in vivo, the in vitro experiments describe a completely
different effect, where AaHNF-4C is slightly upregulat-
ed with a 4 h ecdysone pulse, placing it as a secondary
20E response gene; AaHNF-4B is only upregulated in
the presence of Chx, suggesting a transcript stabiliza-
tion effect of this protein inhibitor; and, finally, the
very surprising result for AaHNF-4A. We observed a
strong upregulation after incubation in culture medium
for 4–16 h (Fig. 4C). In our laboratory, it has been
demonstrated that the blood-meal-dependent signal
that triggers the transcriptional activation of AaVg is
regulated by both the 20E regulatory pathway and an
amino acid dependent pathway [90,91]. Hansen et al.
[90] demonstrated that the TOR pathway is indeed

undetermined [72]. Several lines of evidence have sug-
gested a role for vertebrate ERRa and ERRc in the
control of metabolic genes. Both isoforms are involved
in the regulation of hepatic pyruvate metabolism,
specifically inhibiting glycolytic flux through regulation
of key enzymes in the oxidation of glucose to acetyl-
CoA. This family of NRs act synergistically with the
peroxisome proliferator-activated receptor c coactiva-
tor (PGC1-a) and forkhead transcription factor ( FoxO1),
blocking the conversion of pyruvate to acetyl-CoA in
the mitochondria, while insulin suppresses its effect
[96].
AaTll mRNA was highly expressed in newly eclosed
female fat bodies, sharply decreased to minimum levels
by day 4 pre-vitellogenesis, and remained low during
the whole vitellogenic period, with only small non-sig-
nificant fluctuations observed (Fig. 3E). Such a lack of
fluctuation with known ecdysone titers in vivo is in
agreement with the lack of effect of any in vitro treat-
ment on its expression levels (data not shown). In
D. melanogaster, Tll NR acts as a gap gene during the
early steps of embryogenesis and is involved in con-
trolling terminal genes that result in normal develop-
ment of head and posterior structures [97,98]. Later in
embryonic development, Tll is also necessary for the
establishment of the D. melanogaster embryonic visual
system as well as for the development of the most
anterior region of the brain [99,100]. Tlx, the verte-
brate ortholog of Tll, is required for the correct devel-
opment of the visual system and neurogenesis

not shown). Furthermore, it has been shown that both
proteins were reported to act as repressors of the
AaEcR ⁄ AaUSP heterodimer through protein–protein
binding with AaUSP [13], thus preventing formation
of the functional ecdysone receptor and consequently
inhibiting AaVg expression. AaHR38 sequesters
AaUSP during the pre-vitellogenic period while AaSvp
operates during the termination period [13].
In summary, this study provides a general over-
view of the complete family of A. aegypti NR genes
expressed during the vitellogenic period, a period
important not only for the events that will provide
nourishment for the developing oocyte, but also for a
wide variety of metabolic processes. A. aegypti is an
anautogenous mosquito and an extremely efficient dis-
ease vector because it requires host contact. This
understanding of the molecular regulation of vitello-
genesis is important to achieve significant advances in
the development of future vector- and vector-borne,
disease-control strategies.
Experimental procedures
Annotation of A. aegypti NRs
A set of amino acid sequences corresponding to the 21
NRs identified in D. melanogaster (FlyBase Source; http://
flybase.bio.indiana.edu) were used to search for orthologs
in A. aegypti (http://aaegypti.vectorbase.org/index.php)
using the BLASTP tool against the predicted protein data-
set of A. aegypti. The sequences gleaned from the
A. aegypti database were examined manually through mul-
tiple sequence alignment using clustalw against previously

Expression of identified NRs in the fat body and
ovaries of adult females following a blood meal
To determine whether the identified and annotated NRs are
expressed in a temporal manner in two reproductive tissues
of adult female A. aegypti, RT-PCR analysis was conducted
against abdominal walls with adhering fat bodies (hereafter
referred to as the fat body) and ovaries before vitellogenesis
(pre-vitellogenic), at its onset (6 + 12 h PBM) and at its
peak (18 + 24 h PBM).
Total RNA was extracted from fat bodies and ovaries of
10 females using the TRIzol method (Invitrogen, Carlsbad,
CA, USA). The isolated total RNA was subsequently
cleaned using RNeasy
Ò
mini kit columns (Qiagen, Valencia,
CA, USA), which included an on-column DNase I diges-
tion (Qiagen). cDNA was synthesized from 2.5 lg of the
DNase I-treated RNA using Superscript II (Invitrogen).
To standardize RT-PCR inputs, a master mix containing
HotStarTaq PCR Master Mix (Qiagen) and forward and
reverse primers (final concentration ⁄ PCR = 100 nm each;
see Table S2 for primer sequences) was prepared and ali-
quoted; to this, cDNA of the different tissues and time
points was added. The samples were subjected to PCR
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1249
amplification with a number of cycles within the linear
range of amplification, preincubation at 95 °C for 15 min
followed by 30–40 cycles, depending on the NR (95 °C for
30 s, 60 °C for 30 s, 72 °C for 30 s) and a final elongation

within the fat body, the dissected fat bodies were incubated
for 30 min in culture medium without hormone, followed
by a 6 h incubation in the presence or absence of the
hormone (10
)6
m 20E). To test the effect of the protein
synthesis inhibitor Chx on the expression of previously
uncharacterized A. aegypti NRs and 20E primary response
genes, the fat bodies were pretreated with culture medium
containing 10
)5
m Chx for 30 min, then incubated with
Chx either with or without 20E for an additional 6 h. A
second experiment addressed the effect of an initial induc-
tion by 10
)6
m 20E followed by removal of the said hor-
mone; this was accomplished by first providing 10
)6
m 20E
for 4 h, followed by washing the fat bodies three times with
hormone-free medium and maintaining the fat bodies in
hormone-free media for an additional 12 h. The compara-
ble control was the maintenance of fat bodies for 16 h in
media with or without 10
)6
m 20E. As a control for all
experiments, the fat bodies were incubated in hormone-free
medium supplemented with 10% ethanol (the 20E carrier).
Samples from three biological replicates were analyzed

lation of insect metamorphosis. Receptor 3, 203–209.
7 Sullivan AA & Thummel CS (2003) Temporal profiles
of nuclear receptor gene expression reveal coordinate
transcriptional responses during Drosophila develop-
ment. Mol Endocrinol 17, 2125–2137.
8 Li TR & White KP (2003) Tissue-specific gene expres-
sion and ecdysone-regulated genomic networks in
Drosophila. Dev Cell 5, 59–72.
9 Beckstead RB, Lam G & Thummel CS (2005) The geno-
mic response to 20–hydroxyecdysone at the onset of
Drosophila metamorphosis. Genome Biol 6, R99.
10 Yao TP, Forman BM, Jiang Z, Cherbas L, Chen JD,
McKeown M, Cherbas P & Evans RM (1993) Func-
tional ecdysone receptor is the product of EcR and
Ultraspiracle genes. Nature 366, 476–479.
11 Koelle MR, Talbot WS, Segraves WA, Bender MT,
Cherbas P & Hogness DS (1991) The Drosophila EcR
Nuclear receptors in Aedes aegypti J. Cruz et al.
1250 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
gene encodes an ecdysone receptor, a new member of
the steroid receptor superfamily. Cell 67, 59–77.
12 King-Jones K & Thummel CS (2005) Nuclear recep-
tors – a perspective from Drosophila. Nat Rev Genet 6,
311–323.
13 Zhu J, Miura K, Chen L & Raikhel AS (2003) Cyclic-
ity of mosquito vitellogenic ecdysteroid-mediated sig-
naling is modulated by alternative dimerization of the
RXR homologue Ultraspiracle. Proc Natl Acad Sci
USA 100, 544–549.
14 Martin D, Wang SF & Raikhel AS (2001) The vitello-

lium castaneum. Insect Biochem Mol Biol 38, 430–439.
22 Bonneton F, Chaumot A & Laudet V (2008) Annota-
tion of Tribolium nuclear receptors reveals an increase
in evolutionary rate of a network controlling the ecdy-
sone cascade. Insect Biochem Mol Biol 38, 416–429.
23 Li C, Kapitskaya MZ, Zhu J, Miura K, Segraves W &
Raikhel AS (2000) Conserved molecular mechanism
for the stage specificity of the mosquito vitellogenic
response to ecdysone. Dev Biol 224, 96–110.
24 Lavorgna G, Ueda H, Clos J & Wu C (1991) FTZ-F1,
a steroid hormone receptor-like protein implicated in
the activation of fushi tarazu. Science 252, 848–851.
25 Lavorgna G, Karim FD, Thummel CS & Wu C (1993)
Potential role for a FTZ-F1 steroid receptor superfam-
ily member in the control of
Drosophila metamorpho-
sis. Proc Natl Acad Sci USA 90, 3004–3008.
26 Laudet V, Hanni C, Coll J, Catzeflis F & Stehelin D
(1992) Evolution of the nuclear receptor gene super-
family. EMBO J 11, 1003–1013.
27 Laudet V (1997) Evolution of the nuclear receptor
superfamily: early diversification from an ancestral
orphan receptor. J Mol Endocrinol 19, 207–226.
28 Escriva H, Langlois MC, Mendonca RL, Pierce R &
Laudet V (1998) Evolution and diversification of the
nuclear receptor superfamily. Ann NY Acad Sci 839,
143–146.
29 The Nuclear Receptors Committe (1999) A unified
nomenclature system for the nuclear receptor super-
family. Cell 97, 161–163.

37 Kapitskaya MZ, Li C, Miura K, Segraves W & Raik-
hel AS (2000) Expression of the early-late gene encod-
ing the nuclear receptor HR3 suggests its involvement
in regulating the vitellogenic response to ecdysone in
the adult mosquito. Mol Cell Endocrinol 160, 25–37.
38 Carney GE & Bender M (2000) The Drosophila
ecdysone receptor (EcR) gene is required maternally
for normal oogenesis. Genetics 154, 1203–1211.
39 Cherbas L, Hu X, Zhimulev I, Belyaeva E & Cherbas
P (2003) EcR isoforms in Drosophila: testing
J. Cruz et al. Nuclear receptors in Aedes aegypti
FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS 1251
tissue-specific requirements by targeted blockade and
rescue. Development 130, 271–284.
40 Cruz J, Mane-Padros D, Belles X & Martin D (2006)
Functions of the ecdysone receptor isoform-A in the
hemimetabolous insect Blattella germanica revealed by
systemic RNAi in vivo. Dev Biol 297, 158–171.
41 Buszczak M, Freeman MR, Carlson JR, Bender M,
Cooley L & Segraves WA (1999) Ecdysone response
genes govern egg chamber development during mid-
oogenesis in Drosophila. Development 126, 4581–4589.
42 Terashima J & Bownes M (2006) E75A and E75B have
opposite effects on the apoptosis ⁄ development choice
of the Drosophila egg chamber. Cell Death Differ 13,
454–464.
43 Swevers L, Eystathioy T & Iatrou K (2002) The
orphan nuclear receptors BmE75A and BmE75C of the
silkmoth Bombyx mori: hormonal control and ovarian
expression. Insect Biochem Mol Biol 32, 1643–1652.

inducible gene responsible for the 75B early puff in
Drosophila encodes two new members of the steroid
receptor superfamily. Genes Dev 4, 204–219.
52 Zhou B, Hiruma K, Jindra M, Shinoda T, Segraves
WA, Malone F & Riddiford LM (1998) Regulation of
the transcription factor E75 by 20-hydroxyecdysone
and juvenile hormone in the epidermis of the tobacco
hornworm, Manduca sexta, during larval molting and
metamorphosis. Dev Biol 193, 127–138.
53 Ashburner M (1974) Sequential gene activation by
ecdysone in polytene chromosomes of
Drosophila
melanogaster. II. The effects of inhibitors of protein
synthesis. Dev Biol 39, 141–157.
54 Hurban P & Thummel CS (1993) Isolation and charac-
terization of fifteen ecdysone-inducible Drosophila
genes reveal unexpected complexities in ecdysone regu-
lation. Mol Cell Biol 13, 7101–7111.
55 Horner MA, Chen T & Thummel CS (1995) Ecdyster-
oid regulation and DNA binding properties of
Drosophila nuclear hormone receptor superfamily
members. Dev Biol 168, 490–502.
56 Hiruma K & Riddiford LM (2001) Regulation of
transcription factors MHR4 and betaFTZ-F1 by
20-hydroxyecdysone during a larval molt in the tobacco
hornworm, Manduca sexta. Dev Biol 232, 265–274.
57 Hiruma K & Riddiford LM (2007) The coordination of
the sequential appearance of MHR4 and dopa decarbox-
ylase during the decline of the ecdysteroid titer at the
end of the molt. Mol Cell Endocrinol 276, 71–79.

early late puff contains E78, an ecdysone-inducible
gene that encodes a novel member of the nuclear hor-
mone receptor superfamily. Cell 75, 307–320.
Nuclear receptors in Aedes aegypti J. Cruz et al.
1252 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS
66 Russell SR, Heimbeck G, Goddard CM, Carpenter AT
& Ashburner M (1996) The Drosophila Eip78C gene is
not vital but has a role in regulating chromosome
puffs. Genetics 144, 159–170.
67 Ayer S, Walker N, Mosammaparast M, Nelson JP,
Shilo BZ & Benyajati C (1993) Activation and repres-
sion of Drosophila alcohol dehydrogenase distal tran-
scription by two steroid hormone receptor superfamily
members binding to a common response element.
Nucleic Acids Res 21, 1619–1627.
68 Ohno CK & Petkovich M (1993) FTZ-F1 beta, a novel
member of the Drosophila nuclear receptor family.
Mech Dev 40, 13–24.
69 Niimi T, Morita S, Yamashita O & Yaginuma T
(1997) The profiles of mRNA levels for BHR39, a
Bombyx homolog of Drosophila hormone receptor 39,
and Bombyx FTZ-F1 in the course of embryonic devel-
opment and diapause. Dev Gene Evol 207, 410–412.
70 Huet F, Ruiz C & Richards G (1995) Sequential gene
activation by ecdysone in Drosophila melanogaster: the
hierarchical equivalence of early and early late genes.
Development 121, 1195–1204.
71 Ohno CK, Ueda H & Petkovich M (1994) The Dro-
sophila nuclear receptors FTZ-F1 alpha and FTZ-F1
beta compete as monomers for binding to a site in the

Woodard CT (1999) The Drosophila beta FTZ-F1
orphan nuclear receptor provides competence for
stage-specific responses to the steroid hormone
ecdysone. Mol Cell 3, 143–149.
80 Lam G & Thummel CS (2000) Inducible expression of
double-stranded RNA directs specific genetic interfer-
ence in Drosophila. Curr Biol 10, 957–963.
81 Raikhel AS & Lea AO (1991) Control of follicular
epithelium development and vitelline envelope forma-
tion in the mosquito; role of juvenile hormone and
20-hydroxyecdysone. Tissue Cell 23, 577–591.
82 Sun GC, Hirose S & Ueda H (1994) Intermittent
expression of BmFTZ-F1, a member of the nuclear
hormone receptor superfamily during development of
the silkworm Bombyx mori. Dev Biol 162, 426–437.
83 Kageyama Y, Masuda S, Hirose S & Ueda H (1997)
Temporal regulation of the mid-prepupal gene FTZ-
F1: DHR3 early late gene product is one of the plural
positive regulators. Genes Cells 2, 559–569.
84 Sladek FM, Zhong WM, Lai E & Darnell JE Jr (1990)
Liver-enriched transcription factor HNF-4 is a novel
member of the steroid hormone receptor superfamily.
Genes Dev 4, 2353–2365.
85 Drewes T, Senkel S, Holewa B & Ryffel GU (1996)
Human hepatocyte nuclear factor 4 isoforms are
encoded by distinct and differentially expressed genes.
Mol Cell Biol 16, 925–931.
86 Navas MA, Munoz-Elias EJ, Kim J, Shih D & Stoffel
M (1999) Functional characterization of the MODY1
gene mutations HNF4(R127W), HNF4(V255M), and

receptors. Genes Dev 8, 538–553.
93 Heard DJ, Norby PL, Holloway J & Vissing H (2000)
Human ERRgamma, a third member of the estrogen
receptor-related receptor (ERR) subfamily of orphan
nuclear receptors: tissue-specific isoforms are expressed
during development and in the adult. Mol Endocrinol
14, 382–392.
94 Giguere V (2002) To ERR in the estrogen pathway.
Trends Endocrinol Metab 13, 220–225.
95 Horard B & Vanacker JM (2003) Estrogen receptor-
related receptors: orphan receptors desperately seeking
a ligand. J Mol Endocrinol 31, 349–357.
96 Zhang Y, Ma K, Sadana P, Chowdhury F, Gaillard S,
Wang F, McDonnell DP, Unterman TG, Elam MB &
Park EA (2006) Estrogen-related receptors stimulate
pyruvate dehydrogenase kinase isoform 4 gene expres-
sion. J Biol Chem 281, 39897–39906.
97 Steingrimsson E, Pignoni F, Liaw GJ & Lengyel JA
(1991) Dual role of the Drosophila pattern gene tailless
in embryonic termini. Science 254, 418–421.
98 Hulskamp M & Tautz D (1991) Gap genes and gradi-
ents – the logic behind the gaps. Bioessays 13, 261–268.
99 Rudolph KM, Liaw GJ, Daniel A, Green P, Courey
AJ, Hartenstein V & Lengyel JA (1997) Complex regu-
latory region mediating tailless expression in early
embryonic patterning and brain development. Develop-
ment 124, 4297–4308.
100 Younossi-Hartenstein A, Green P, Liaw GJ, Rudolph
K, Lengyel J & Hartenstein V (1997) Control of early
neurogenesis of the Drosophila brain by the head gap

in mosquito oocytes. Roux’s Arch Dev Biol 199,
114–121.
109 Lea AO (1964) Studies on the dietary and endocrine
regulation of autogenous reproduction in Aedes tae-
niorhynchus (Wied.). J Med Entomol 39, 40–44.
110 Raikhel AS, Deitsch KW & Sappington TW (1997)
Culture and analysis of the insect fat body. In The
Molecular Biology of Insect Dissease Vectors. A Meth-
ods Manual (Crampton JM, Beard CB & Louis C,
eds), pp. 507–522. Chapman and Hall, London.
111 Hagedorn HH, O’Connor JD, Fuchs MS, Sage B, Sch-
laeger DA & Bohm MK (1975) The ovary as a source
of alpha-ecdysone in an adult mosquito. Proc Natl
Acad Sci USA 72, 3255–3259.
Supporting information
The following supplementary material is available:
Table S1. NRs of Drosophila melanogaster, Aedes
aegypti, Anopheles gambiae, Apis mellifera and
Tribolium castaneum.
Table S2. Primers used in RT-PCR and real-time
PCR.
This supplementary material can be found in the
online version of this article.
Please note: Wiley-Blackwell is not responsible for
the content or functionality of any supplementary
materials supplied by the authors. Any queries (other
than missing material) should be directed to the corre-
sponding author for the article.
Nuclear receptors in Aedes aegypti J. Cruz et al.
1254 FEBS Journal 276 (2009) 1233–1254 ª 2009 The Authors Journal compilation ª 2009 FEBS


Nhờ tải bản gốc

Tài liệu, ebook tham khảo khác

Music ♫

Copyright: Tài liệu đại học © DMCA.com Protection Status