ArticlePDF Available

G-protein-coupled receptor participates in 20-hydroxyecdysone signaling on the plasma membrane

Authors:

Abstract and Figures

Animal steroid hormones are conventionally known to initiate signaling via a genomic pathway by binding to the nuclear receptors. The mechanism by which 20E initiates signaling via a nongenomic pathway is unclear. We illustrate that 20E triggered the nongenomic pathway through a plasma membrane G-protein-coupled receptor (named ErGPCR) in the lepidopteran insect Helicoverpa armigera. The transcript of ErGPCR was increased at the larval molting stage and metamorphic molting stage by 20E regulation. Knockdown of ErGPCR via RNA interference in vivo blocked larval-pupal transition and suppressed 20E-induced gene expression. ErGPCR overexpression in the H. armigera epidermal cell line increased the 20E-induced gene expression. Through ErGPCR, 20E modulated Calponin nuclear translocation and phosphorylation, and induced a rapid increase in cytosolic Ca2+ levels. The inhibitors of T-type voltage-gated calcium channels and canonical transient receptor potential calcium channels repressed the 20E-induced Ca2+ increase. Truncation of the N-terminal extracellular region of ErGPCR inhibited its localization on the plasma membrane and 20E-induced gene expression. ErGPCR was not detected to bind with the steroid hormone analog [3H]Pon A. These results suggest that ErGPCR participates in 20E signaling on the plasma membrane.
Content may be subject to copyright.
R E S E A R C H Open Access
G-protein-coupled receptor participates in
20-hydroxyecdysone signaling on the plasma
membrane
Mei-Juan Cai
, Du-Juan Dong
, Yu Wang, Peng-Cheng Liu, Wen Liu, Jin-Xing Wang and Xiao-Fan Zhao
*
Abstract
Background: Animal steroid hormones are conventionally known to initiate signaling via a genomic pathway by
binding to the nuclear receptors. The mechanism by which 20E initiates signaling via a nongenomic pathway is
unclear.
Results: We illustrate that 20E triggered the nongenomic pathway through a plasma membrane G-protein-coupled
receptor (named ErGPCR) in the lepidopteran insect Helicoverpa armigera. The transcript of ErGPCR was increased at the
larval molting stage and metamorphic molting stage by 20E regulation. Knockdown of ErGPCR via RNA interference
in vivo blocked larvalpupal transition and suppressed 20E-induced gene expression. ErGPCR overexpression in the H.
armigera epidermal cell line increased the 20E-induced gene expression. Through ErGPCR, 20E modulated Calponin
nuclear translocation and phosphorylation, and induced a rapid increase in cytosolic Ca
2+
levels. The inhibitors of
T-type voltage-gated calcium channels and canonical transient receptor potential calcium channels repressed the
20E-induced Ca
2+
increase. Truncation of the N-terminal extracellular region of ErGPCR inhibited its localization on
the plasma membrane and 20E-induced gene expression. ErGPCR was not detected to bind with the steroid hormone
analog [
3
H]Pon A.
Conclusion: These results suggest that ErGPCR participates in 20E signaling on the plasma membrane.
Keywords: Steroid hormones, G-protein-coupled receptors, Protein phosphorylation, Calcium influx, Signal transduction
Background
Animal steroid hormones are lipid-soluble molecules
conventionally known to initiate signaling via a genomic
pathway. Steroid hormones enter the nucleus by freely
diffusing through cell membranes to combine with intra-
cellular nuclear receptors for gene transactivation. Nuclear
receptors act by forming homodimers or heterodimers.
For example, the glucocorticoid receptor [1] and estrogen
receptor [2] form a homodimer, and insect ecdysone
receptor (EcR) forms a heterodimer with an ultraspiracle
protein (USP), the ortholog of retinoid X receptor in ver-
tebrates [3]. The heat shock proteins Hsp90 and Hsp70
interact with the nuclear receptors to facilitate their DNA
binding activity in fruit flies [4] and mammals [5]. Hsp90
[6] and Hsc70 [7] have been found to be involved in insect
steroid hormone signaling by differential interaction with
the nuclear receptors. However, plant steroid hormones,
such as brassinosteroids, employ a nongenomic pathway
to initiate signaling by combining with plasma membrane
receptor kinases for gene transactivation [8]. The bras-
sinosteroids exhibit structural similarity to the steroid
hormones of vertebrates and insects [9]. These studies
suggest the existence of similar pathways in plants and
animals, which should be further studied.
Previous studies indicated that animal steroid hormones
can trigger nongenomic actions through the cytoplasmic
membrane [10]. For example, estrogen activates phos-
phoinositide 3 kinase to recruit protein kinase B to the
membrane in mammals [11]. A G-protein-coupled seven
transmembrane receptor (GPR30) can act as the membrane
receptor for estrogen [12], and it has been renamed G-pro-
tein-coupled estrogen receptor 1 (GPER) [13]. However,
* Correspondence: xfzhao@sdu.edu.cn
Equal contributors
The Key Laboratory of Plant Cell Engineering and Germplasm Innovation,
Ministry of Education, Shandong Provincial Key Laboratory of Animal Cells
and Developmental Biology, School of Life Sciences, Shandong University,
Jinan 250100, Shandong, China
© 2014 Cai et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain
Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article,
unless otherwise stated.
Cai et al. Cell Communication and Signaling 2014, 12:9
http://www.biosignaling.com/content/12/1/9
GPR30 is located in the endoplasmic reticulum [14] and
may be translocated to the plasma membrane [15]. New
studies have suggested that GPER is ubiquitinated at the
cell surface, and constitutively internalized in an arrestin-
independent manner. Moreover, GPER does not recycle to
the plasma membrane [16]. GPER acts as a stand-alone
membrane receptor of pregenomic action independent
on the estrogen nuclear receptor [17]. GPER also func-
tions in the nervous system, and may be a pharmaceutical
target [18].
In insects, the ATP-binding cassette transporter E23
can act as a general negative regulator of ecdysteroid
signaling by transporting 20E outside of the cell [19]. 20E
promotes neuroblast proliferation during metamorphosis
partly by suppressing nitric oxide production in <15 min
without protein synthesis or transcription [20], and the cell
membrane receptor of 20E is assumed to be a leucine-rich
repeat receptor kinase [21]. The plasma membrane of the
anterior silk gland of Bombyx mori binds [
3
H] ponasterone
A([
3
H]Pon A), suggesting that the anterior silk gland may
express an unknown membrane 20E receptor [22]. 20E in-
duces intracellular Ca
2+
release into the cytoplasm via an
unknown G-protein-coupled receptor (GPCR) pathway in
the anterior silk gland of silkworms [23]. The Drosophila
dopamine receptor DmDopEcR binds [
3
H]Pon A, and is
considered as a 20E membrane receptor [24]. Ecdysteroids
trigger rapid Ca
2+
increase, including intracellular Ca
2+
release, and extracellular Ca
2+
influx through GPCR in
mouse skeletal muscle cells [25]. In our previous study,
we demonstrated that 20E regulates the rapid nuclear
translocation and phosphorylation of Calponin for gene
expression in Helicoverpa armigera [26]. These findings
suggest that 20E has membrane receptors and a nonge-
nomic signaling pathway.
In this study, we reported an ecdysone-responsible
GPCR (ErGPCR) participates in 20E signaling on the
plasma membrane. The knockdown of ErGPCR disrupted
several biological processes, including the larvalpupal
metamorphosis, expression of 20E-induced genes, subcel-
lular translocation and phosphorylation of Calponin, and
20E-induced cytosolic Ca
2+
increase.
Results
ErGPCR is involved in 20E-regulated gene expression
It has been known that 20E regulates the gene expression
of the nuclear receptor EcRB1 and transcription factors
Br,USP1,E75B,andHR3 [27]. Suramin disrupts GPCR
binding with the G protein by blocking the association of
Gproteinαand βγ subunits [28]. Suramin is widely used
to study GPCR- and G-protein-initiated cell signaling,
including the 20E-induced GPCR pathway in the anterior
silk gland of silkworms [23], cytosolic Ca
2+
increase, and
protein kinase C activation [29]. Thus, the involvement of
GPCRs in 20E-induced gene expression was analyzed
using the GPCR inhibitor suramin in a lepidopteran H.
armigera epidermal cell line (HaEpi cell line, established
in our laboratory) [30]. 20E significantly promoted the
expression of EcRB1,BrZ2,HHR3,andUSP1 compared
with the DMSO solvent control. However, the 20E-induced
transcript increase was repressed by the addition of
suramin (Figure 1). These results suggest that GPCRs
are probably involved in 20E-regulated mRNA levels.
We identified six GPCR candidates from the expressed
sequence tags (EST) of the cDNA library of the HaEpi
cell line using BLASTX assay (http://www.ncbi.nlm.nih.
gov/) (Additional file 1: Figure S1, Table S1). The mRNA
levels of six GPCR candidates were upregulated by 20E
induction, and two non-GPCR ESTs were unaffected.
Knockdown of No. 16666 and ErGPCR in the HaEpi
cells using RNA interference (RNAi) decreased EcRB1,
BrZ2,HHR3,andUSP1 transcript levels in 20E induction.
The knockdown of the other four GPCR candidates
affected one to three 20E-induced gene transcripts
(Additional file 1: Figure S2). These results suggest the
involvement of GPCRs in 20E-induced gene expression.
ErGPCR was further studied regarding its expression
profile during development. The deduced amino acid
sequence of ErGPCR contains a signal peptide at the
N-terminus and seven transmembrane domains (Additional
file 1: Figure S3). ErGPCR belongs to methuselah-like
proteins in the class B secretin GPCR family based on
NCBI Blast analysis (http://blast.ncbi.nlm.nih.gov/Blast.
cgi). ErGPCR has 57% identity with Spodoptera frugiperda
GPCR, 32% with Tribolium castaneum GPCR, and 30%
with Drosophila melanogaster GPCR (Additional file 1:
Figure 1 Involvement of GPCRs in the 20E pathway in HaEpi cells
as determined by quantitative real-time reverse transcription
polymerase chain reaction (qRT-PCR) analysis. DMSO treatment
was used as the solvent control for 20E. DMSO plus suramin 50 μM
treatment for 1 h was used to determine the toxic effects of suramin
on the cells. The HaEpi cells were pretreated with 50 μM suramin for
1 h and then exposed to 1 μM 20E for another 6 h. The results are
based on the ΔΔCT calculation by normalization of the β-actin
gene. Error bars represent the standard deviation of three
independent replicates. Asterisks indicate significant differences
(Studentsttest, *p<0.05).
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 2 of 16
http://www.biosignaling.com/content/12/1/9
Figure S4). However, D. melanogaster DmDopEcR, Homo
sapiens GPR30, and H. sapiens beta-2 adrenergic receptor
(AR) are not found by BLASTX analysis. This finding sug-
gests that ErGPCR is less similar to DmDopEcR, GPR30,
and AR. Phylogenetic analysis indicated that ErGPCR does
not cluster with DmDopEcR, GPR30, and AR. These
results illustrate that these GPCRs belong to different
GPCR groups (Additional file 1: Figure S5).
The transcript level of ErGPCR was increased at the
larval molting stage (5 M) and metamorphic molting stage
(sixth-instar 72 h larvae to pupae) in the tissues (Figure 2).
Given that the 20E titer is higher during molting and
metamorphosis in lepidopteran insect Manduca sexta
[27], the hormone induction on the mRNA levels of
ErGPCR was examined. The ErGPCR transcript level was
upregulated in the midgut from 3 h to 24 h after 20E
injection into the sixth-instar larvae. JH III injection
into the sixth-instar larvae did not affect the ErGPCR
transcript levels, but repressed the 20E-induced upreg-
ulation of ErGPCR (Figure 3). These data suggest that
ErGPCR mRNA level is upregulated by 20E signaling.
To confirm that 20E upregulates ErGPCR, we knocked
down the nuclear receptor of 20E, EcRB1, and analyzed
the transcript of ErGPCR.WhenEcRB1 was knocked
down, the upregulation of ErGPCR induced by 20E was
blocked (Additional file 1: Figure S6). These results reveal
that 20E upregulates ErGPCR transcript via the nuclear
receptor EcRB1.
Figure 2 The increased transcripts of ErGPCR during molting
and metamorphosis. (A),(B) and (C) ErGPCR is highly expressed
during molting and metamorphosis in epidermis, midgut and fat
body detected by qRT-PCR. 5Fis the fifth instar 12 h larvae; 5Mis
the fifth instar molting larvae; 60to6120 h are the 6th instar
larvae in hours; p 0 to p 8 are the pupae in days.
Figure 3 Hormonal induction of ErGPCR in the midgut by
injection of 20E (A) or JH III (B) or 20E +JH III (C) into the 6th
instar 6 h larvae (500 ng/larva) analyzed by qRT-PCR. DMSO is a
solvent control. β-actin gene was used as the quantitative control
for the mRNA. The asterisks indicate significant differences between
20E treatment and DMSO solvent control by studentsttest analysis
from three independent repeats (*p< 0.05, n = 3).
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 3 of 16
http://www.biosignaling.com/content/12/1/9
ErGPCR is involved in the larvalpupal transition in vivo
by regulating gene expression
The function of ErGPCR in larvalpupal transition was
determined through RNAi by injecting dsErGPCR into the
larval hemocoel. The knockdown of ErGPCR blocked lar-
valpupal transition (Figure 4A). In the dsRNA of green
fluorescent protein (dsGFP)-injected control, 90% of the
larvae pupated, whereas 10% died. However, in dsErGPCR
treatment, only 29% of the larvae pupated, 50% died, and
21% displayed larvalpupal chimeras (Figure 4B). Of the
29% that pupated after ErGPCR knockdown, the duration
of development was significantly delayed compared with
the dsGFP control: a 23 h delay from fifth instar to the
sixth instar, and a 52 h delay from the sixth instar to the
pupal stage (Figure 4C). RTPCR showed that ErGPCR
was significantly knocked down by four consecutive
Figure 4 ErGPCR knockdown by dsRNA injection blocks larval-pupal transition. (A) Phenotypes after ErGPCR knockdown (dsErGPCR injection
into the 5th instar larval hemocoel). Scale bar = 1 cm. (B) Statistical analysis of the phenotype in A.(C) 50 h to 60 h: 0 h of the 5th instar to 0 h
of the 6th instar 0 h; 60 h to pupation: 0 h of the 6th instar to pupation. (D) The efficiency of ErGPCR knockdown, analyzed by semi-quantitative
RT-PCR. (E) The gene transcripts in the larval epidermis after ErGPCR knockdown were determined by qRT-PCR analysis. Error bars represent the
standard deviations of three replicates. Asterisks denote significant differences (*p< 0.05; **p< 0.01, via Studentsttest) based on 30 larval
samples with three replicates.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 4 of 16
http://www.biosignaling.com/content/12/1/9
dsErGPCR injections into the larvae (Figure 4D). The
transcript levels of the genes involved in the 20E pathway,
including EcRB1,USP1,HHR3,BrZ2,andE75B,werede-
creased in the larval epidermis after ErGPCR knockdown
(Figure 4E). These results suggest that ErGPCR is related
to larvalpupal transition and gene expression in vivo.
ErGPCR is located on the membrane and is necessary for
20E-induced gene expression
Immunocytochemical analysis using rabbit anti-ErGPCR
polyclonal antibodies showed that ErGPCR was located
on the plasma membrane of HaEpi cells. The green
fluorescence of ErGPCR overlapped with the red-stained
cell membrane to display an orange color by confocal laser
scanning microscopy (Figure 5A). Moreover, the overex-
pression of ErGPCR-GFP (C-terminal with GFP) was also
located on the cell plasma membrane (Figure 5B). ErGPCR
knockdown in the HaEpi cells decreased the 20E-induced
transcript levels of EcRB1,USP1,HHR3,BrZ2,andE75B
compared with the dsGFP-treated control (Figure 5C).
By contrast, ErGPCR overexpression led to an increase
in the transcript levels of EcRB1,BrZ2,HHR3,andUSP1
(Figure 5D). Moreover, 20E upregulated the mRNA level
of insulator body protein mod(mdg4)1a not through
EcRB1 but through ErGPCR (Figures 5E and F). These
results suggest that ErGPCR may serve as the initiation
step of 20E signal amplification on the plasma membrane
as nongenomic or pregenomic action before the hierarch-
ical control of 20E-induced genomic action.
20E regulates protein nuclear translocation and
phosphorylation via ErGPCR
The nongenomic pathway is characterized by rapid protein
translocation and phosphorylation [31]. The Calponin
protein has been demonstrated to undergo quick nuclear
translocation and phosphorylation by 20E induction [26].
Thus, ErGPCR was knocked down in the HaEpi cells to de-
termine its function in 20E-regulated rapid translocation
and phosphorylation of Calponin. Calponin was mainly lo-
calized in the cytoplasm in the DMSO-negative control
cells, but was translocated into the nucleus after 20E induc-
tion. However, after ErGPCR knockdown, 20E could not
induce the nuclear translocation of Calponin (Figure 6A).
Moreover, the 20E-mediated Calponin phosphorylation was
suppressed when ErGPCR was silenced (Figure 6B). The
protein synthesis inhibitor anisomycin did not inhibit
20E-induced Calponin phosphorylation (Figure 6C). These
results suggest that 20E regulates Calponin nuclear trans-
location and phosphorylation via ErGPCR.
20E regulates cellular Ca
2+
release and influx via ErGPCR
to regulate gene expression
The increase in cellular Ca
2+
is another characteristic of
the nongenomic pathway of steroid hormones [32]. Thus,
ErGPCR was knocked down in the HaEpi cells to deter-
mine the function of ErGPCR in the rapid 20E-regulated
Ca
2+
increase. When the cells were incubated in calcium-
free buffer (DPBS), cytosolic Ca
2+
level increased rapidly
by 20E treatment, and peaked at approximately 50 s, then
declined to a lower level at 120 s. Following the addition
of 1 mM calcium into DPBS at 120 s, the cytosolic Ca
2+
levels gradually increased and then remained constant.
However, suramin (50 μM) pretreatment for 1 h inhibited
the 20E-induced rapid increase in cytosolic Ca
2+
levels
(Figure 7A). When ErGPCR was knocked down by RNAi,
the 20E-induced Ca
2+
increase, including intracellular Ca
2+
release, and extracellular Ca
2+
influx, was also inhibited
compared with the control (Figure 7B). These findings
suggest that 20E induces rapid intracellular Ca
2+
release
and extracellular Ca
2+
influx via ErGPCR.
Various calcium channel blockers, including the T-type
voltage-gated calcium channel inhibitor flunarizine dihy-
drochloride (FL), L-type calcium channel inhibitor verap-
amil hydrochloride (Ve) [33], transient receptor potential
(TRP) calcium channel store-operated channel (SOC)
inhibitor 2-aminoethoxydiphenyl borate (2-APB) [34],
and receptor-operated (ROC) TRPC3 channel inhibitor
pyrazole (Pyr3) [35], were employed to determine the
involvement of calcium channels in 20E-induced extracellu-
lar Ca
2+
influx. The 20E-induced Ca
2+
influx was restrained
by 50 μM FL without affecting intracellular Ca
2+
release.
By contrast, 20E-induced Ca
2+
release and influx were un-
affected by 100 μM Ve (Figure 7C). The 2-APB inhibitor
(10 μMto100μM) had no effect on the 20E-induced Ca
2+
release and influx. However, 10 μM Pyr3 suppressed the
20E-induced Ca
2+
influx, but had no effect on intracellular
Ca
2+
release (Figure 7D). These results reveal that T-type
calcium channels and TRPC3 channels are involved in
20E-induced Ca
2+
flux.
To investigate the effect of the cellular Ca
2+
increase
on the 20E-induced gene expression and 20E-induced
Calponin phosphorylation, we performed qRTPCR and
western blot. The 20E-induced upregulation of EcRB1,
BrZ2,HHR3,andUSP1 was suppressed by FL and Pyr3
(Figure 7E). Meanwhile, the 20E-induced phosphorylation
of Calponin was inhibited (Figure 7F). By contrast, Ve and
2-APB inhibitors had no effect on the 20E-induced gene
expression and 20E-induced Calponin phosphorylation.
These results show that the 20E-induced rapid intracellu-
lar Ca
2+
increase is required for 20E-regulated gene ex-
pression and protein phosphorylation.
To examine the mechanism by which 20E regulates
gene expression through ErGPCR and Ca
2+
signaling,
ChIP experiments were performed by anti-RFP antibody
in the EcRB1-RFP-overexpressing HaEpi cells. In M. sexta,
one ecdysone response element (EcRE1, GGGGTCAATG
AACCG) was identified in 20E reporter gene hormone
receptor 3 (HR3). 20E regulates EcRB1/USP1 heterodimer
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 5 of 16
http://www.biosignaling.com/content/12/1/9
Figure 5 ErGPCR is located on the plasma membrane and is involved in 20E-induced gene expression in HaEpi cells. (A) Green fluorescence
indicated ErGPCR protein detected with anti-ErGPCR (antibody specificity is shown in Additional file 1: Figure S7) and ALEXA 488labeled
goat anti-rabbit secondary antibody by confocal microscope. The nuclei were stained with 4-6-diamidino-2-phenylindole dihydrochloride
(DAPI). (B) Green fluorescence protein (GFP, green) alone was detected in the entire cell; the plasma membrane (red) was marked with
1,1-dioctadecyl-3,3,3,3-tetramethylindocarbocyanine perchlorate (DiI); and the orange color are the overlapping of ErGPCR-GFP (green)
and membrane (red) by confocal microscope. Scale bar =20 μm. (C) and (D) The effects of ErGPCR knockdown and overexpression on
20E-induced gene expression, analyzed by qRT-PCR. (E) and (F) 20E upregulated mod(mdg4)1a expression not through EcRB1 but through
ErGPCR, by qRT-PCR analysis. β-actin was used as the quantitative control. * indicates significant differences (p< 0.05) among the treatments
via Studentsttest from three independent replicates.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 6 of 16
http://www.biosignaling.com/content/12/1/9
binding to EcRE to regulate gene transcription [36].
We cloned the 5' regulatory region of Helicoverpa HR3
(HHR3) that contains putative EcRE (GGGGTCAA
TGAACTG), which has one Tdifferent from EcRE1 in
MHR3. The EcRE from HHR3 is proven to be active by
GFP-plasmid examination. Fewer PCR product (EcRE)
Figure 6 Knockdown of ErGPCR by RNAi blocks 20E-induced nuclear translocation and phosphorylation of Calponin in the HaEpi cells.
(A) Green fluorescence represents Calponin detected with anti-Calponin antibody. Blue fluorescence indicates DAPI-stained nuclei. The image in the
white box shows the efficacy of ErGPCR knockdown (by dsRNA incubation for 24 h followed by 1 μM 20E induction for 1 h), determined by RT-PCR; N,
C, and dsG indicate ErGPCR transcripts in normal cells, dsGFP-treated cells, and dsErGPCR-treated cells, respectively. (B) 20E-induced phosphorylation of
Calponin was detected with anti-Calponin antibody by western blot analysis.Cal-P indicates phosphorylated Calponin. (C) The effect of anisomycin on
20E-induced phosphorylation of Calponin was analyzed using anti-His monoclonal antibody by western blot analysis. Cal-GFP (56 kDa) is the fused
expressed Calponin with GFP in the HaEpi cells; Cal-GFP-P indicates phosphorylated Calponin-GFP; GFP (35 kDa) was expressed alone in the cells as a
control. The cells were incubated with 10 μM anisomycin for 1 h and then treated with DMSO or 1 μM 20E for 1 h.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 7 of 16
http://www.biosignaling.com/content/12/1/9
was detected from the immunoprecipitates in the pIEx-4-
RFP-transfected control samples after various treatments
by anti-RFP antibody, because the RFP recognized by
anti-RFP antibody did not bind to DNA. By contrast, in
the pIEx-4-EcRB1-RFP-transfected cells, the PCR product
(EcRE) was detected from the immunoprecipitates in 20E
induction by anti-RFP antibody. However, after ErGPCR
knockdown, the PCR product (EcRE) was significantly
decreased compared with the dsGFP treatment control
(Figure 8A). These results suggest that 20E via ErGPCR
regulates EcRB1 binding to EcRE to regulate the 20E-
induced gene transcription.
Similarly, in the pIEx-4-RFP-transfected control samples,
fewerPCRproduct(EcRE)wasdetectedintheimmuno-
precipitate by anti-RFP antibody after various treatments.
By contrast, in the pIEx-4-EcRB1-RFP-transfected samples,
the PCR product containing EcRE was detected in 20E
induction by anti-RFP antibody, which recognized the
binding of EcRB1-RFP to EcRE. However, fewer DNA
product containing EcRE was detected after the addition
of inhibitors FL and Pyr3. Ve and 2-APB had no effect
on the PCR product (EcRE) (Figure 8B). These results
suggest that 20E via cellular Ca
2+
signaling modulates the
binding of EcRB1 to EcRE, and regulates the 20E-induced
gene transcription.
N-terminal extracellular region is essential to ErGPCR
function in the 20E signaling pathway
Truncated mutations of ErGPCR in the N-terminal region
(aa 20 to 197) (OVErGPCR
Δ20197 aa
) or second inner loop
Figure 7 ErGPCR is involved in the 20E-induced increase in cytosolic Ca
2+
levels. (A) Suramin inhibited the 20E-induced Ca
2+
increase (1 μM
20E, 1 mM calcium chloride, 50 μM suramin). F represents the fluorescence of cells after treatment, whereas F
0
denotes the average fluorescence of
cells before treatment. Fluorescence was detected every 6 s for 360 s using a Laser Scan Confocal Microscope Carl Zeiss LSM 700 (Thornwood, NY,
USA) at 555 nm and then analyzed using the Image Pro-Plus software. (B) ErGPCR knockdown using dsRNA (5 μg/mL) incubation inhibited the
20E-induced Ca
2+
increase. (C) The T-type calcium channel blocker flunarizine dihydrochloride (FL, 50 μM) inhibited the 20E-indued Ca
2+
influx. The
L-type calcium channel blocker verapamil hydrochloride (Ve, 100 μM) did not affect the 20E-induced Ca
2+
influx. (D) The transient receptor potential
(TRP) channel blocker pyrazole (Pyr3, 10 μM) inhibited the 20E-induced Ca
2+
increase. 2-Aminoethoxydiphenyl borate (2-APB) (10 μMto100μM) did
not affect 20E-induced Ca
2+
influx. (E) qRT-PCR showing the involvement of Ca
2+
signal in 20E-induced gene expression. Cells were incubated in 1 μM
20E for 6 h after different inhibitors pretreatment for 1 h and the RNA was isolated for qRT-PCR. β-actin was used as the quantitative control. *indicates
significant differences (p< 0.05) among the treatments via Studentsttest from three independent replicates. (F) Western blot showing the
involvement of Ca
2+
signal in 20E-induced Calponin phosphorylation. pIEx-4-Cal-GFP was overexpressed in HaEpi cells for 48 h. The cells were
treated with 1 μM 20E for 1 h after different inhibitors pretreatment for 1 h. The GFP was overexpressed as controls.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 8 of 16
http://www.biosignaling.com/content/12/1/9
(aa 275 to 308) (OVErGPCR
Δ275308 aa
) were overexpressed
by fusing with GFP to analyze the functional domain of
ErGPCR in the 20E signaling pathway (Figure 9A). Over-
expressed full-length ErGPCR (OVErGPCR) was located
ontheplasmamembrane.However,theN-terminal
region-truncated ErGPCR (OVErGPCR
Δ20197 aa
)was
in the cytoplasm, whereas the second inner loop truncation
(OVErGPCR
Δ275308 aa
) was located in both the plasma
membrane and cytoplasm (Figures 9B and C). Correlated
with its location, the truncation of the N-terminal extra-
cellular region of ErGPCR (OVErGPCR
Δ20197 aa
)caused
a decrease in the 20E-induced transcript levels of EcRB1,
BrZ2,HHR3,andUSP1 compared with the full-length
OVErGPCR. By contrast, truncation of the second inner
loop of ErGPCR (OVErGPCR
Δ275308 aa
)didnotaffect
the 20E-induced transcript levels of EcRB1,BrZ2,HHR3,
and USP1 (Figure 9D). These results indicate that the
complete structure of ErGPCR is essential to its plasma
membrane location, and the N-terminal extracellular
region is required for ErGPCR function in the 20E sig-
naling pathway.
ErGPCR is not detected to bind with the 20E analog
[
3
H]Pon A
ErGPCR and EcRB1 were overexpressed in HaEpi cells by
fusing with GFP at the C-terminus (Figure 10A) for the
binding experiments. No increase in [
3
H]Pon A (0.1 nM)
was detected with the increase in cell numbers (1 × 10
4
cells to 100 × 10
4
cells) in ErGPCR-GFP-transfected cells
compared with the normal cells, GFP-overexpressed cells,
or EcRB1-GFP-overexpressed cells (Figure 10B). The
increase in [
3
H]Pon A was not detected in the plasma
membrane fractions (5 μgto500μg) from HaEpi cells
that overexpressed ErGPCR-GFP compared with that
in the membrane fractions from the normal cells and
GFP-overexpressed cells (Figure 10C). These results sug-
gest that cells or cell membrane fractions could bind with
[
3
H]Pon A in a cell- or cell membrane fraction-dependent
manner. However, overexpression of ErGPCR does not
increase [
3
H]Pon A binding by this analysis.
Discussion
Although studies have shown that GPCRs are involved
in 20E signaling, definitive evidence of this involvement
is scarce. Our study reveals that ErGPCR regulates 20E
signaling on the plasma membrane. Through ErGPCR,
20E regulates gene expression, fast protein translocation
and phosphorylation, rapid intracellular Ca
2+
increase, and
larvalpupal transition.
20E regulates genomic action through the ErGPCR-mediated
nongenomic pathway
20E initiates the genomic pathway by binding with its
nuclear hormone receptor EcR to regulate gene expression
for metamorphosis [37]. 20E upregulated the mRNA levels
of EcRB1,USP1,HHR3,BrZ2,andE75B.Knockdownof
ErGPCR repressed the binding of EcRB1 to EcRE thus
blocked 20E-induced expression of EcRB1,USP1,HHR3,
BrZ2,andE75B in the cell line and larvae, which resulted
in blocking the 20E genomic pathway, thereby inhibiting
metamorphosis. These results indicate that 20E initiates a
nongenomic pathway to regulate a 20E-mediated genomic
pathway via ErGPCR. In positive feedback, ErGPCR tran-
script was upregulated by 20E via EcRB1. 20E does not act
through EcRB1 to upregulate the mRNA level of the insula-
tor body protein mod(mdg4)1a in HaEpi cells [38]. Instead,
mod(mdg4)1a is upregulated by 20E through ErGPCR.
These results suggest the existence of various pathways
in 20E signaling. The reason that knockdown of EcRB1
repressed ErGPCR but did not repress mod(mdg4)1a
Figure 8 ChIP analysis showing ErGPCR regulates 20E-induced
EcRB1 binding to EcRE through cellular Ca
2+
signaling. (A) ErGPCR
knockdown suppresses the EcRB1 binding to EcRE under 20E treatment.
Cells were transfected with pIEx-4-RFP or pIEx-4-EcRB1-RFP for 24 h. The
cells were treated by dsErGPCR or dsGFP for 12 h, then exposed in 1
μM 20E or DMSO for 6 h. (B) FL and Pyr3 repress EcRB1 binding to
EcRE in 20E treatment. Cells were transfected with pIEx-4-RFP or
pIEx-4-EcRB1-RFP for 48 h. The cells were subjected in 1 μM20Eor
DMSO for 6 h after different calcium channel inhibitors pretreatment
for 1 h. ChIP was carried out without antibody (negative control) or
with anti-RFP antibody. The DNA fragment (EcRE) was amplified by
qRT-PCR using HHR3F/R primers. Input is the positive control of
nonimmunoprecipitated chromatin.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 9 of 16
http://www.biosignaling.com/content/12/1/9
might because the time difference of mRNA transcription
and protein translation of ErGPCR.
Steroid hormones, such as mammalian estrogen and
insect ecdysone, are conventionally thought to exert
their actions through binding to intracellular receptors
because of their small molecules and lipid solubility.
However, growing evidence indicates that steroid hor-
mones also exert rapid cell surface-initiated actions by
binding to membrane receptors [39], such as the estro-
gen membrane receptor GPR30 [14]. Rapid protein
subcellular translocation and phosphorylation (within
minutes) are the outcomes of a nongenomic signaling
pathway [24]. 20E regulates the rapid nuclear trans-
location and phosphorylation of Calponin for gene trans-
activation in H. armigera [26]. We found that 20E
regulated Calponin nuclear translocation and subse-
quent phosphorylation through ErGPCR. This finding
suggests that 20E functions in the membrane via a
nongenomic pathway to regulate protein translocation
and phosphorylation, which may contribute to the activa-
tion of transcription factors and formation of transcription
complexes.
Figure 9 The N-terminus of ErGPCR is the key domain for its cell membrane localization and function. (A) mutation sites of ErGPCR,
20-197 indicates the truncation of amino acids 20197; 275-308 indicates the truncation of amino acids 275308. (B) The localization of the
overexpressed GFP (OVGFP), full length of ErGPCR fused with GFP (OVErGPCR-GFP), truncation of N-terminal extracellular region of ErGPCR fused
with GFP (OVErGPCR
20-197aa
-GFP), and truncation of the second inner loop of ErGPCR fused with GFP (OVErGPCR
276-308aa
-GFP) in the HaEpi cells
by confocal microscope. (C) Subcellular localization of ErGPCR and two mutants were confirmed by western blot with His-tag antibody. (D) The
effects of the mutations of ErGPCR on the transcripts of EcRB1,BrZ2,HHR3 and USP1 induced by 20E were examined by qRT-PCR, with β-actin as
control. Bars indicate mean ± S.D. from three independent experiments. Asterisk indicates significant difference (p < 0.05).
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 10 of 16
http://www.biosignaling.com/content/12/1/9
ErGPCR is involved in 20E-increased cytosolic Ca
2+
levels
20E increases the cytosolic Ca
2+
levels by promoting
the release of Ca
2+
from the intracellular endoplasmic
reticulum via an unknown GPCR in silkworm silk glands
[23]. 20E also regulates Ca
2+
influx from extracellular
sources via an unknown GPCR that activates calcium
channels in murine skeletal muscles [25]. Voltage-gated
calcium channels are essential in regulating extracellular
Ca
2+
influx in a wide variety of tissues [40]. T-type Ca
2+
channels are involved in 20E-induced nuclear and DNA
fragmentation in silkworm silk glands [23]. GPCRs
serve as chaperones and interact with voltage-gated cal-
cium channels to form complexes [41]. Our data show
that 20E regulates rapid intracellular Ca
2+
release and
extracellular Ca
2+
influx through ErGPCR, and T-type
voltage-gated Ca
2+
channels are involved in Ca
2+
influx.
In addition, we found that the 20E-induced Ca
2+
influx
was also inhibited by the TRP channel inhibitor Pyr3. This
result suggests that TRP channels are also involved in 20E-
induced Ca
2+
influx. TRP channels are non-voltage-gated
Ca
2+
channels involved in Ca
2+
entry [42]. TRP channels
are classified into six subfamilies according to their primary
structure and function, including ROC and SOC [43].
GPCRs directly or indirectly modulate several TRP chan-
nels [44,45]. TRP channels are associated with steroid hor-
mones in mammals [46]. Rapid calcium release or influx in
the cells is the outcome of nongenomic signaling. Calcium
is an important secondary messenger that regulates numer-
ous essential physiologic processes, including protein kinase
C activation, for further protein phosphorylation [47] and
gene transcription. In our study, when the cellular Ca
2+
was
blocked by inhibitors, 20E-induced gene expression and the
phosphorylation of Calponin were blocked. These findings
confirm the function of calcium on gene expression and
protein phosphorylation as the secondary messenger, and
reveal that 20E regulates the cellular calcium via ErGPCR
to regulate the genomic pathway.
ErGPCR does not bind with the steroid hormone
analog [
3
H]Pon A
In classical GPCR signaling pathways, ligands bind to
cell surface transmembrane receptors, such as the β2 ARs,
and cause conformational changes in their transmembrane
and intracellular domains [48]. Numerous studies have
reported the binding of several GPCRs with 20E, such
as the binding of DmDopEcR with [
3
H]Pon A [24], or an
unknown GPCR in the anterior silk gland of silkworms
binding with [
3
H]Pon A [22]. However, we did not detect
the binding of ErGPCR with [
3
H]Pon A using the whole
cells and cell membrane fractions by overexpressing
ErGPCR in HaEpi cells. Thus, ErGPCR is likely transiently
activated by 20E without any stable ligand binding.
Based on NCBI Blast analysis (http://blast.ncbi.nlm.nih.
gov/Blast.cgi), ErGPCR belongs to methuselah-like pro-
teins in the class B secretin GPCR family, but DmDopEcR
shows homology with vertebrate ARs [24]. Identification
and phylogenetic analysis using amino acid sequences
show that ErGPCR differs from GPR30, beta-2 AR, or
Drosophila DmDopEcR, which may explain the differences
in ligand binding activity. Another possibility is the
analytical method, which needs further study in next work.
GPR30 has shown negligible binding to estrogen (17β-
estradiol) in several studies [49], which may be due to
the different analytical methods [17]. A major challenge
in the study of steroid hormone nongenomic pathways
is the binding assay of GPCR with the steroid hormone
[50]. Although ErGPCR did not bind ecdysteroid in our
study, this result is of particular importance in many
cellular responses to 20E, including 20E-induced mRNA
levels, protein subcellular translocation and phosphoryl-
ation, and cellular Ca
2+
increase. Whether other GPCRs
can bind with 20E needs further exploration.
A total of 800 GPCRs have been discovered in mammals
[51], 1000 in Caenorhabditis elegans [52], and 200 in D.
melanogaster [53]. GPCRs involved in steroid membrane
Figure 10 Binding assay of [
3
H]Pon A to whole cells or membrane fractions. (A) Western blot analysis of the overexpressed GFP, ErGPCR-
GFP and EcRB1-GFP. (B) Binding of [
3
H]Pon A to the whole HaEpi cells that expressed GFP, ErGPCR-GFP or EcRB1-GFP, respectively. The cells (1100 ×
10
4
) were incubated with 0.1 nM [
3
H]Pon A (5740 cpm) for 1 h at 27°C, respectively. (C) The membrane fractions (5, 50, 250, 500 μgmembranepro-
teins) isolated from above cells were incubated with 0.1 nM [
3
H]Pon A for 1 h at 27°C, respectively. cpm: counts per minute of [
3
H]Pon A. The data
marked with different small letters indicate a significant difference at 0.05 levels by the one-way ANOVA with Duncans test.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 11 of 16
http://www.biosignaling.com/content/12/1/9
signaling may differ among various organisms. GPCRs
form homodimers, heterodimers, oligomers, or complexes
in the signalsomes of the membrane [54]. The mechanism
underlying GPCRs in the 20E membrane pathway requires
further study.
Conclusions
ErGPCR participates in 20E-regulated gene expression,
rapid Calponin nuclear translocation and phosphorylation,
rapid intracellular Ca
2+
release, and extracellular Ca
2+
in-
flux via T-type calcium channels and TRP channels. The
N-terminal extracellular region is critical for the function
of ErGPCR in the 20E signaling pathway. ErGPCR is
necessary for the larvalpupal transition in H. armigera
development (Figure 11).
Methods and materials
Insect
Cotton boll worms, H. armigera, were reared in our
laboratory on an artificial diet of wheat, soybean, vitamins,
and inorganic salts under a 14 h light:10 h dark cycle at
27°C.
Quantitative real-time reverse-transcriptase PCR (qRT-PCR)
Approximately 5 μg of the total RNA from each sample
was reverse transcribed into first-strand cDNA for qRT-
PCR analysis (First Strand cDNA Synthesis Kit, Sangon,
China). qRT-PCR was performed using SsoFastEvaGreen
Supermix (BIO-RAD, Shanghai, China). Thermocycling
(94°C for 20 s, 58°C for 20 s, and 72°C for 20 s) was
performed for 40 cycles using the CFX96real-time sys-
tem (BIO-RAD). The experiment was repeated three
times using three independent RNA samples for statistical
analysis. β-actin was used as the cDNA quality and
quantity controls. The primers used for qRT-PCR are
listed in Additional file 1: Table S2.
Suramin inhibition
H. armigera epidermal cells (HaEpi) cells were cultured
until 80% confluence at 27°C in 25 cm
2
culture flasks
using Graces medium (Invitrogen, Carlsbad, CA, USA)
supplemented with 10% fetal bovine serum (FBS, Mdgenics,
St. Louis, MO, USA). The cells were incubated with 50 μM
suramin (sodium salt; Sigma Chemical, St. Louis, MO,
USA) for 1 h and then exposed to 1 μM20Eforanother
6 h. Gene expression was determined via qRT-PCR.
Screening of the target GPCR by qRT-PCR
dsRNA was produced using a MEGAscriptRNAi kit
(Ambion, Austin, TX, USA). Some GPCR ESTs obtained
through random sequencing of the HaEpi cells, a cell line
previously established in our laboratory, were individually
knocked down using RNA interference. Lipofectamine
2000 (Invitrogen, Carlsbad, CA, USA) was used for dsRNA
transfection. Briefly, 5 μgofdsRNAand8μL of Lipofecta-
mine 2000 were added to 125 μL of FBS-free Graces
medium incubated at room temperature for 30 min. The
reagents were mixed and incubated for another 20 min,
Figure 11 Diagram showing the ErGPCR-mediated nongenomic pathway response to 20E. 20E through ErGPCR nongenomic pathway
regulates the cytosolic calcium fast increase, including the release of the intracellular Ca
2+
, and the influx of the extracellular Ca
2+
via T-type
calcium channels and TRP channels. 20E also through ErGPCR regulates the rapid phosphorylation and nuclear translocation of Calponin, and
through ErGPCR regulates gene transcription in the genomic pathway for metamorphosis.
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 12 of 16
http://www.biosignaling.com/content/12/1/9
and then directly added into HaEpi cells in a six-well plate
containing 0.8 mL of Graces medium per well. After incu-
bation at 27°C for 24 h, the cells were rinsed and then re-
fed with fresh medium containing 10% FBS. The cells
were cultured with 1 μM 20E for 6 h. Control cells were
treated with the same amount of dsGFP. Total RNA was
then extracted from the cells for qRT-PCR based on three
independent replicates.
Cloning of full-length cDNA of ErGPCR
We obtained an EST with the 3end of ErGPCR by
random sequencing of the cDNA library of the insect
during metamorphosis. The 5end of the gene was
amplified via PCR using the gene-specific reverse pri-
mer ErGPCRF1 and the 5primer through the Genome
Walker method as described by Clontech Laboratories
Inc. (cat. no. 638904; Clontech, CA, USA).
Recombinant expression of ErGPCR in Escherichia coli and
antiserum preparations
The ErGPCR fragment was amplified using the primers
ErGPCRExpF and ErGPCRExpR. The PCR product was
cloned into pET30a plasmid, expressed in Escherichia
coli rosette cells, and then cultured in a LuriaBertani
medium (1% tryptone, 0.5% yeast extract, 1% NaCl, and
25 μg/mL kanamycin). The target protein was purified
using His-bind resin to produce polyclonal rabbit anti-
serum.Thespecificityoftheantibodywasdetermined
via western blot analysis using horseradish peroxidase
labeled goat anti-rabbit polyclonal secondary antibodies
(Zhongshan, Beijing).
Immunocytochemistry
The cells grown on cover slips were fixed with 4% parafor-
maldehyde in phosphate-buffered saline (PBS; 140 mM
NaCl, 2.7 mM KCl, 10 mM Na
2
HPO
4
, 1.8 mM KH
2
PO
4
,
pH 7.4) for 10 min. The fixed cells were incubated with
0.2% Triton-X 100 in PBS for 8 min, blocked with 2% bo-
vine serum albumin (BSA) in PBS for 30 min, and then in-
cubated with primary antibody against the target gene
(1:100 dilution in 2% BSA/PBS) overnight at 4°C. The cells
were washed and then incubated with the ALEXA 488
labeled goat anti-rabbit secondary antibodies (diluted
1:1000 in 2% BSA/PBS) for 1 h at 37°C. Nuclei were
stained with DAPI (1 μg/mL in PBS) for 10 min. Fluores-
cence was detected using a Laser Scan Confocal Microscope
Carl Zeiss LSM 700 (Thornwood, NY, USA).
ErGPCR overexpression and truncated mutation of ErGPCR
PCR was used to prepare truncated mutations of ErGPCR.
ErGPCR fragments were amplified via PCR with various
primers (Additional file 1: Table S2) using proofreading
DNA polymerase. The mutated ErGPCR was amplified via
PCR using the ErGPCR fragments as templates. The open
reading frame of ErGPCR and different mutated ErGPCRs
were inserted into the pIEx-4 plasmid (Merck, Darmstadt,
Germany), fused with GFP. The plasmid was transfected
into HaEpi cells with Cellfectin following the protocol of
the supplier (Invitrogen, Carlsbad, CA, USA). Afterward,
20E was added to the cells at a final concentration of
1μM. An equal volume of DMSO was used as the
solvent control for 20E. DiI (1,1-dioctadecyl-3,3,3,3-tetra-
methylindocarbocyanine perchlorate; Beyotime, Shanghai,
China) was used for plasma membrane staining.
Examination of Calponin translocation and phosphorylation
Subcellular Calponin translocation and phosphorylation
were detected by immunocytochemistry and immunoblot-
ting using rabbit polyclonal antibodies against Helicoverpa
Calponin. After ErGPCR knockdown, the cells were treated
with 1 μM 20E for 0.5 h to 3 h. Control cells were treated
via the same method using GFP dsRNA. Fluorescence was
detected using an Olympus BX51 fluorescence microscope.
The phosphorylation analysis was performed by western
blot.
Calcium ion detection
HaEpi cells were seeded and cultured for 72 h in a six-well
tissue culture plate with 10% FBS Gracesmediumat27°C.
The cells were incubated with dsRNA for 24 h as previously
described. The cells were incubated in a 3 μMAMester
Calcium Crimsondye (Invitrogen, Carlsbad, CA, USA) in
Graces medium for 30 min at 27°C. The cells were then
washed with DPBS (2.7 mM KCl, 1.5 mM KH
2
PO
4
,and
8mMNa
2
HPO
4
)andexposedto1μM20EinDPBS
for 2 min for detection of intracellular calcium release.
Afterward, 1 mM calcium chloride was added to induce
extracellular calcium influx. Fluorescence was detected at
555 nm every 6 s for 360 s using a Laser Scan Confocal
Microscope Carl Zeiss LSM 700 (Thornwood, NY, USA).
Data were analyzed using the Image Pro-Plus software.
For the inhibition experiments, the cells were pretreated
with different inhibitors for 1 h prior to 20E treatment.
The GPCR inhibitor suramin, T-type voltage-gated calcium
channel inhibitor flunarizine dihydrochloride, L-type
calcium channel inhibitor verapamil hydrochloride, and
TRP channel inhibitors 2-APB and Pyr3 were purchased
from Sigma Chemical (St. Louis, MO, USA).
Chromatin immunoprecipitation (ChIP)
The HaEpi cells were seeded in a six-well plate. Cells
were transfected with pIEx-4-EcRB1-RFP at a density
of 2 × 10
6
. After 24 h, the cells were transfected with
dsErGPCR, and the controls were incubated with
dsGFP. After 24 h, the cells were subjected to either
DMSO or 1 μM 20E. After 6 h, the cells were cross-linked
with 0.5% formaldehyde at 37°C for 10 min, followed by
quenching at 0.125 M glycine at room temperature for
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 13 of 16
http://www.biosignaling.com/content/12/1/9
10 min. The cells were then washed with ice-cold 1 × PBS
and harvested at 6,000 rpm for 5 min. Cells were re-
suspended with SDS lysis buffer (1% SDS, 10 mM EDTA,
50 mM TrisHCl, pH 8.1) and sonicated to yield average
DNA fragments of 200 bp to 1000 bp. After centrifugation
to remove cell debris, the lysates were pre-cleared with
protein A resin at 4°C for 1 h, followed by incubation with
no antibody (negative control) or anti-RFP antibody over-
night. Immunoprecipitated protein-DNA complexes were
incubated with protein A for an additional 2 h at C. The
complexes were washed with low-salt buffer [0.1% SDS,
1.0% Triton X-100, 2 mM EDTA, 200 mM TrisHCl
(pH 8.0), 150 mM NaCl] once, high-salt wash buffer [0.1%
SDS, 1.0% Triton X-100, 2 mM EDTA, 20 mM TrisHCl
(pH 8.0), 500 mM NaCl] once, LiCl wash buffer [10 mM
TrisHCl (pH 8.1), 0.25 M LiCl, 1 mM EDTA, 1% NP-40,
1% deoxycholate] once, and TE buffer [10 mM TrisHCl
(pH 8.1), 1 mM EDTA] two times. The bound proteins
were eluted with elution buffer (1% SDS, 0.1 M NaHCO
3
).
DNA-protein crosslinks were reversed at 65°C overnight,
followed by RNase and proteinase K treatment. DNA was
purified with phenol/chloroform and ethanol precipita-
tion, and analyzed by qRTPCR using HHR3F/R primers
(Additional file 1: Table S2). The negative control cells
were transfected with the same volume of pIEx-4-RFP,
and the cells received the same treatment as above.
RNAi in larvae
T7 promoter-containing PCR primers (GPCRRNA-iF,
GPCRRNA-iR, GFPRNAiF and GFPRNAiR in Additional
file 1: Table S2) were used to amplify the gene fragments.
The PCR product purified with phenolchloroform was
used as a template to synthesize dsRNA using the MEGA-
script RNAi Kit, as previously described. The dsRNA was
diluted in nuclease-free water to 0.4 μg/μL. Afterward,
5μL was injected into the fifth instar larvae at 6 h and
at 30 h, as well as into the sixth instar at 6 h and 30 h. The
controls were injected with dsGFP. Three independent
experiments were performed using 30 larvae each.
[
3
H]Pon A binding assays
Cell membranes that express ErGPCR, EcRB1 and GFP
were prepared from HaEpi cells with the plasmid
ErGPCR-GFP-pIEx-4, EcRB1-GFP-pIEx-4, and GFP-pIEx-4.
The details are as follows: cells were collected by centrifu-
gation (1700×g, 10 min, 4°C) and then resuspended in
15 mL of HEPES buffer [20 mM HEPES, 6 mM MgCl
2
,
1 mM ethylene diamine tetraacetic acid (EDTA), 1 mM
ethylene glycol bis (2-aminoethyl) tetraacetic acid (EGTA),
pH 7.4]. After sonication, the homogenate was centrifuged
at 1700×g for 10 min. The resulting supernatant was cen-
trifuged at 48000×g for 1 h at 4°C. The pellet was resus-
pended in HEPES buffer, and the protein concentration
was determined via the Bradford method. For the binding
assay, a range of membrane fractions were incubated with
1nM[
3
H]Pon A (Perkin Elmer, MA, USA) at 27°C for 1 h
in 200 μL of binding buffer (20 mM HEPES, 100 mM
NaCl, 6 mM MgCl
2
, 1 mM EDTA, 1 mM EGTA). For the
saturation experiments, reaction mixtures containing
50 μg of the membrane fraction were incubated at 27°C
for 1 h in the presence of the appropriate [
3
H]Pon A con-
centration in the binding buffer. Nonspecific binding was
determined in the presence of 1 μM 20E. After incubation,
particulate proteins were collected on glass fiber filters.
The filters were then added to 5 mL of scintillation fluid.
Radioactivity was determined using a SN-6930 liquid scin-
tillation counter (Shanghai Hesuo Rihuan Photoelectric
Instrument Co., Ltd., China). The whole cell binding ex-
periments used the same method but without sonication
and membrane preparation.
Additional file
Additional file 1: The data sets supporting the results of this
article are included within the article. Figure S1. Alignment of the
GPCR ESTs obtained by random sequencing the Helicoverpa epidermal
cell line. Figure S2. Screen of the target GPCR involved in 20E-induced
gene expression by qRT-PCR. Figure S3. Nucleotide and deduced amino
acid sequence of ErGPCR. Figure S4. Multiple alignments of ErGPCR with
other G-protein-coupled receptors from different insects or vertebrates.
Figure S5. Phylogenetic analysis of ErGPCR. Figure S6. 20E upregulates
ErGPCR through EcRB1.Figure S7. The recombinant expression of ErGPCR
fragments in E. coli.Table S1. Identification of the GPCRs. Table S2.
Primers used in dsRNA synthesis and qRT-PCR.
Abbreviations
20E: 20-hydroxyecdysone; JH: Juvenile hormone; GPCR: G protein-coupled re-
ceptor; EcRB1: Ecdysone nuclear receptor B1; USP1: Ultraspiracle protein 1;
HR3: Hormone receptor 3; BrZ2: Broad isoform Z2; DMSO: Dimethylsulfoxide;
dsRNA: Double-stranded RNA; RNAi: RNA interference; GFP: Green fluorescent
protein; RFP: Red fluorescent protein; HaEpi: An epidermal cell line from
Helicoverpa armigera; FBS: Fetal bovine serum; DAPI: 4-6-diamidino-2-
phenylindole dihydrochloride; BSA: Bovine serum albumin; TRP: Transient
receptor potential; SOC: Store-operated channel; ROC: Receptor-operated
channel; DiI: 1,1-dioctadecyl-3,3,3,3-tetramethylindocarbocyanine
perchlorate; 2-APB: 2-aminoethoxydiphenyl borate; Cpm: Counts per minute
of [
3
H] Pon A; qRT-PCR: quantitative reverse transcription polymerase chain
reaction; ChIP: Chromatin immunoprecipitation; EcRE: Ecdysone response
element.
Competing interests
The authors declare no conflict of interests.
Authorscontributions
Mei-Juan Cai performed the overexpression of ErGPCR and Ca
2+
detection.
Du-Juan Dong constructed the pIEx-4-ErGPCR-GFP plasmid. Yu Wang cloned
the gene. Wen Liu overexpressed ErGPCR in cell line. Peng-Cheng Liu examined
the phosphorylation and translocation of Calponin. Jin-Xing Wang directed the
research. Xiao-Fan Zhao designed the studies and wrote the manuscript. All
authors read and approved the final manuscript.
Acknowledgments
This work was supported by the grants from the National Natural Science
Foundation of China (No. 31230067) and the National Basic Research
Program of China (973 Program, No. 2012CB114101) and Ph. D. Programs
Foundation of Ministry of Education of China (No. 20120131110025). The
L4440 and pIEx-4 plasmids and HT115 (DE3) are kindly provided by
Dr. Marek Jindra and Masako Asahina of the Biology Center, Czech Academy
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 14 of 16
http://www.biosignaling.com/content/12/1/9
of Sciences and Department of Molecular Biology, University of South Bohemia,
Czech Republic. We thank professor Lynn Moorhead Riddiford in Janelia Farm
Research Campus, Howard Hughes Medical Institute in United States giving
suggestions on preparing this manuscript.
GenBank number: JQ809653.
Received: 1 November 2013 Accepted: 3 February 2014
Published: 10 February 2014
References
1. Ratman D, Vanden Berghe W, Dejager L, Libert C, Tavernier J, Beck IM, De
Bosscher K: How glucocorticoid receptors modulate the activity of other
transcription factors: a scope beyond tethering. Mol Cell Endocrinol 2013,
380:4154.
2. Parker MG, Arbuckle N, Dauvois S, Danielian P, White R: Structure and
function of the estrogen receptor. Ann N Y Acad Sci 1993, 684:119126.
3. Oro AE, McKeown M, Evans RM: Relationship between the product of the
Drosophila ultraspiracle locus and the vertebrate retinoid X receptor.
Nature 1990, 347:298301.
4. Arbeitman MN, Hogness DS: Molecular chaperones activate the
Drosophila ecdysone receptor, an RXR heterodimer. Cell 2000, 101:6777.
5. Pratt WB, Galigniana MD, Morishima Y, Murphy PJ: Role of molecular
chaperones in steroid receptor action. Essays Biochem 2004, 40:4158.
6. Liu W, Zhang FX, Cai MJ, Zhao WL, Li XR, Wang JX, Zhao XF: The
hormone-dependent function of Hsp90 in the crosstalk between
20-hydroxyecdysone and juv enile hor mone signaling pathways in insects
is determined by differential phosphorylation and protein interactions.
Biochim Biophys Acta 2013, 1830:51845192.
7. Zheng WW, Yang DT, Wang JX, Song QS, Gilbert LI, Zhao XF: Hsc70
binds to ultraspiracle resulting in the upregulation of 20-hydroxyecdsone-
responsive genes in Helicoverpa armigera.Mol Cell Endocrinol 2010,
315:282291.
8. Bishop GJ, Koncz C: Brassinosteroids and plant steroid hormone
signaling. Plant Cell 2002, 14(Suppl):S97110.
9. Mussig C, Altmann T: Brassinosteroid signaling in plants. Trends Endocrinol
Metab 2001, 12:398402.
10. Losel R, Wehling M: Nongenomic actions of steroid hormones. Nat Rev
Mol Cell Biol 2003, 4:4656.
11. Aronica SM, Kraus WL, Katzenellenbogen BS: Estrogen action via the cAMP
signaling pathway: stimulation of adenylate cyclase and cAMP-regulated
gene transcription. Proc Natl Acad Sci U S A 1994, 91:85178521.
12. Prossnitz ER, Maggiolini M: Mechanisms of estrogen signaling and gene
expression via GPR30. Mol Cell Endocrinol 2009, 308:3238.
13. Maggiolini M, Picard D: The unfolding stories of GPR30, a new
membrane-bound estrogen receptor. J Endocrinol 2010, 204:105114.
14. Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER: A
transmembrane intracellular estrogen receptor mediates rapid cell
signaling. Science 2005, 307:16251630.
15. Sanden C, Broselid S, Cornmark L, Andersson K, Daszkiewicz-Nilsson J, Martensson
UE, Olde B, Leeb-Lundberg LM: G protein-coupled estrogen receptor 1/G
protein-coupled receptor 30 localizes in the plasma membrane and traffics
intracellularly on cytokeratin intermediate filaments. Mol Pharmacol 2011,
79:400410.
16. Cheng SB, Quinn JA, Graeber CT, Filardo EJ: Down-modulation of the
G-protein-coupled estrogen receptor, GPER, from the cell surface
occurs via a trans-Golgi-proteasome pathway. J Biol Chem 2011,
286:2244122455.
17. Filardo EJ, Thomas P: Minireview: G protein-coupled estrogen receptor-1,
GPER-1: its mechanism of action and role in female reproductive cancer,
renal and vascular physiology. Endocrinology 2012, 153:29532962.
18. Srivastava DP, Evans PD: GPER 1: trials and tribulations of a membrane
oestrogen receptor. J Neuroendocrinol 2013, 25:12191230.
19. Hock T, Cottrill T, Keegan J, Garza D: The E23 early gene of Drosophila
encodes an ecdysone-inducible ATP-binding cassette transporter
capable of repressing ecdysone-mediated gene activation. Proc Natl Acad
Sci U S A 2000, 97:95199524.
20. Champlin DT, Truman JW: Ecdysteroid coordinates optic lobe
neurogenesis via a nitric oxide signaling pathway. Development 2000,
127:35433551.
21. Thummel CS, Chory J: Steroid signaling in plants and insectscommon
themes, different pathways. Genes Dev 2002, 16:31133129.
22. Elmogy M, Iwami M, Sakurai S: Presence of membrane ecdysone receptor
in the anterior silk gland of the silkworm Bombyx mori. Eur J Biochem
2004, 271:31713179.
23. Manaboon M, Iga M, Iwami M, Sakurai S: Intracellular mobilization of Ca2+ by
the insect steroid hormone 20-hydroxyecdysone during programmed cell
deathinsilkwormanteriorsilkglands.JInsectPhysiol2009, 55:122128.
24. Srivastava DP, Yu EJ, Kennedy K, Chatwin H, Reale V, Hamon M, Smith T,
Evans PD: Rapid, nongenomic responses to ecdysteroids and
catecholamines mediated by a novel Drosophila G-protein-coupled
receptor. J Neurosci 2005, 25:61456155.
25. Gorelick-Feldman J, Cohick W, Raskin I: Ecdysteroids elicit a rapid Ca2+
flux leading to Akt activation and increased protein synthesis in skeletal
muscle cells. Steroids 2010, 75:632637.
26. Liu PC, Wang JX, Song QS XFZ: The Participation of Calponin in the cross
talk between 20-hydroxyecdysone and juvenile hormone signaling
pathways by phosphorylation variation. PLoS One 2011, 6:112.
27. Riddiford LM, Hiruma K, Zhou XF, Nelson CA: Insights into the molecular
basis of the hormonal control of molting and metamorphosis from
Manduca sexta and Drosophila melanogaster. Insect Biochem Mol Biol
2003, 33:13271338.
28. Chung WC, Kermode JC: Suramin disrupts receptor-G protein coupling by
blocking association of G protein alpha and betagamma subunits.
J Pharmacol Exp Ther 2005, 313:191198.
29. Lopez-Lopez R, Langeveld CH, Pizao PE, van Rijswijk RE, Wagstaff J, Pinedo
HM, Peters GJ: Effect of suramin on adenylate cyclase and protein kinase
C. Anticancer Drug Des 1994, 9:279290.
30. Shao HL, Zheng WW, Liu PC, Wang Q, Wang JX, Zhao XF: Establishment of
a new cell line from lepidopteran epidermis and hormonal regulation on
the genes. PLoS One 2008, 3:e3127.
31. Raz L, Khan MM, Mahesh VB, Vadlamudi RK, Brann DW: Rapid estrogen
signaling in the brain. Neurosignals 2008, 16:140153.
32. Vasudevan N, Pfaff DW: Membrane-initiated actions of estrogens in
neuroendocrinology: emerging principles. Endocr Rev 2007, 28:119.
33. So HS, Park C, Kim HJ, Lee JH, Park SY, Lee ZW, Kim HM, Kalinec F, Lim DJ,
Park R: Protective effect of T-type calcium channel blocker flunarizine on
cisplatin-induced death of auditory cells. Hear Res 2005, 204:127139.
34. Kovacs G, Montalbetti N, Simonin A, Danko T, Balazs B, Zsembery A, Hediger
MA: Inhibition of the human epithelial calcium channel TRPV6 by
2-aminoethoxydiphenyl borate (2-APB). Cell Calcium 2012, 52:468480.
35. Kiyonaka S, Kato K, Nishida M, Mio K, Numaga T, Sawaguchi Y, Yoshida T,
Wakamori M, Mori E, Numata T, Ishii M, Takemoto H, Ojida A, Watanabe K,
Uemura A, Kurose H, Morii T, Kobayashi T, Sato Y, Sato C, Hamachi I, Mori Y:
Selective and direct inhibition of TRPC3 channels underlies biological
activities of a pyrazole compound. Proc Natl Acad Sci U S A 2009,
106:54005405.
36. Lan Q, Hiruma K, Hu X, Jindra M, Riddiford LM: Activation of a delayed-early
gene encoding MHR3 by the ecdysone receptor heterodimer EcR-B1-USP-1
but not by EcR-B1-USP-2. Mol Cell Biol 1999, 19:48974906.
37. Bender M, Imam FB, Talbot WS, Ganetzky B, Hogness DS: Drosophila
ecdysone receptor mutations reveal functional differences among
receptor isoforms. Cell 1997, 91:777788.
38. Cai MJ, Liu W, He HJ, Wang JX, Zhao XF: Mod(mdg4) participates in
hormonally regulated midgut programmed cell death during
metamorphosis. Apoptosis 2012, 17:13271339.
39. Thomas P: Rapid steroid hormone actions initiated at the cell surface
and the receptors that mediate them with an emphasis on recent
progress in fish models. Gen Comp Endocrinol 2012, 175:367383.
40. Christel C, Lee A: Ca2 + dependent modulation of voltage-gated Ca2+
channels. Biochim Biophys Acta 2012, 1820:12431252.
41. Altier C: GPCR and Voltage-Gated Calcium Channels (VGCC) Signaling
Complexes. Subcell Biochem 2012, 63:241262.
42. Cioffi DL, Barry C, Stevens T: Store-operated calcium entry channels in
pulmonary endothelium: the emerging story of TRPCS and Orai1. Adv
Exp Med Biol 2010, 661:137154.
43. Harteneck C, Klose C, Krautwurst D: Synthetic modulators of TRP channel
activity. Adv Exp Med Biol 2011, 704:87106.
44. Clapham DE: TRP channels as cellular sensors. Nature 2003, 426:517524.
45. Yekkirala AS: Two to tango: GPCR oligomers and GPCR-TRP channel
interactions in nociception. Life Sci 2013, 92:438445.
46. Wagner TF, Loch S, Lambert S, Straub I, Mannebach S, Mathar I, Dufer M, Lis
A, Flockerzi V, Philipp SE, Oberwinkler J: Transient receptor potential M3
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 15 of 16
http://www.biosignaling.com/content/12/1/9
channels are ionotropic steroid receptors in pancreatic beta cells. Nat
Cell Biol 2008, 10:14211430.
47. Clapham DE: Calcium signaling. Cell 2007, 131:10471058.
48. Rasmussen SG, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, Thian FS,
Chae PS, Pardon E, Calinski D, Mathiesen JM, Shah ST, Lyons JA, Caffrey M,
Gellman SH, Steyaert J, Skiniotis G, Weis WI, Sunahara RK, Kobilka BK: Crystal
structure of the beta2 adrenergic receptor-Gs protein complex. Nature
2011, 477:549555.
49. Otto C, Fuchs I, Kauselmann G, Kern H, Zevnik B, Andreasen P, Schwarz G,
Altmann H, Klewer M, Schoor M, Vonk R, Fritzemeier KH: GPR30 does not
mediate estrogenic responses in reproductive organs in mice. Biol Reprod
2009, 80:3441.
50. Norman AW, Mizwicki MT, Norman DP: Steroid-hormone rapid actions,
membrane receptors and a conformational ensemble model. Nat Rev
Drug Discov 2004, 3:2741.
51. Millar RP, Newton CL: The year in G protein-coupled receptor research.
Mol Endocrinol 2010, 24:261274.
52. Bargmann CI: Neurobiology of the Caenorhabditis elegans genome.
Science 1998, 282:20282033.
53. Adams MD, Celniker SE, Holt RA, Evans CA, Gocayne JD, Scherer SE, Venter
JC: The genome sequence of Drosophila melanogaster. Science 2000,
287:21852195.
54. Moreno JL, Holloway T, Gonzalez-Maeso J: G protein-coupled receptor
heterocomplexes in neuropsychiatric disorders. Prog Mol Biol Transl Sci
2013, 117:187205.
doi:10.1186/1478-811X-12-9
Cite this article as: Cai et al.:G-protein-coupled receptor participates
in 20-hydroxyecdysone signaling on the plasma membrane. Cell
Communication and Signaling 2014 12:9.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Cai et al. Cell Communication and Signaling 2014, 12:9 Page 16 of 16
http://www.biosignaling.com/content/12/1/9
... A GPCR, dopamine/ ecdysteroid receptor (DopEcR), is considered as a 20E cell membrane receptor because it binds 20E to regulate non-genomic effects of ecdysteroids in Drosophila melanogaster (Srivastava et al., 2005). Two GPCRs, ecdysone responsive (Er)GPCR-1 (Cai et al., 2014) and ErGPCR-2 transmit the 20E signal via the nongenomic pathway in Helicoverpa armigera. ErGPCR-2 and DopEcR from H. armigera can bind 20E and function as 20E cell membrane receptors (Kang et al., 2019). ...
... ErGPCR-3 is the fourth GPCR identified to bind and transmit 20E in H. armigera, following ErGPCR-1 (Cai et al., 2014), ErGPCR-2 and DopEcR (Kang et al., 2019). Some differences have been observed in these GPCRs, e.g. ...
Article
Full-text available
Animal steroid hormones initiate signaling by passive diffusion into cells and binding to their nuclear receptors to regulate gene expression. Animal steroid hormones can initiate signaling via G protein-coupled receptors (GPCRs); however, the underlying mechanisms are unclear. Here, we show that a newly discovered ecdysone-responsive GPCR, ErGPCR-3, transmits the steroid hormone 20-hydroxyecdysone (20E) signal by binding 20E and promoting its entry into cells in the lepidopteran insect Helicoverpa armigera . Knockdown of ErGPCR-3 in larvae caused delayed and abnormal pupation, inhibited remodeling of the larval midgut and fat body, and repressed 20E-induced gene expression. Also, 20E induced both the interaction of ErGPCR-3 with G proteins and rapid intracellular increase in calcium, cAMP and protein phosphorylation. ErGPCR-3 was endocytosed by GPCR kinase 2-mediated phosphorylation, and interacted with β-arrestin-1 and clathrin, to terminate 20E signaling under 20E induction. We found that 20E bound to ErGPCR-3 and induced the ErGPCR-3 homodimer to form a homotetramer, which increased 20E entry into cells. Our study revealed that homotetrameric ErGPCR-3 functions as a cell membrane receptor and increases 20E diffusion into cells to transmit the 20E signal and promote metamorphosis.
... The 20E-responsive GPCR (initially designated ErGPCR, and later, ErGPCR-1) [47] and ErGPCR-2 [48] are further revealed in Helicoverpa armigera. ErGPCR-1 expression levels are increased at the molting and metamorphic stages under 20E regulation. ...
... armigera epidermal cell line) increases 20E pathway-related gene expression. 20E induces a rapid increase in cytosolic Ca 2+ levels and promotes calponin nuclear translocation and phosphorylation via ErGPCR-1 [47]. ErGPCR-2 has a similar function to ErGPCR-1, such as regulation of rapid increases in intracellular Ca 2+ levels, and phosphorylation of USP [48]. ...
Article
Full-text available
G protein-coupled receptors (GPCRs) are cell membrane receptors for various ligands. Recent studies have suggested that GPCRs transmit animal steroid hormone signals. Certain GPCRs have been shown to bind steroid hormones, for example, G protein-coupled estrogen receptor 1 (GPER1) binds estrogen in humans, and Drosophila dopamine/ecdysteroid receptor (DopEcR) binds the molting hormone 20-hydroxyecdysone (20E) in insects. This review summarizes the research progress on GPCRs as animal steroid hormone cell membrane receptors, including the nuclear and cell membrane receptors of steroid hormones in mammals and insects, the 20E signaling cascade via GPCRs, termination of 20E signaling, and the relationship between genomic action and the nongenomic action of 20E. Studies indicate that 20E induces a signal via GPCRs to regulate rapid cellular responses, including rapid Ca ²⁺ release from the endoplasmic reticulum and influx from the extracellular medium, as well as rapid protein phosphorylation and subcellular translocation. 20E via the GPCR/Ca ²⁺ /PKC/signaling axis and the GPCR/cAMP/PKA-signaling axis regulates gene transcription by adjusting transcription complex formation and DNA binding activity. GPCRs can bind 20E in the cell membrane and after being isolated, suggesting GPCRs as cell membrane receptors of 20E. This review deepens our understanding of GPCRs as steroid hormone cell membrane receptors and the GPCR-mediated signaling pathway of 20E (20E-GPCR pathway), which will promote further study of steroid hormone signaling via GPCRs, and presents GPCRs as targets to explore new pharmaceutical materials to treat steroid hormone-related diseases or control pest insects. Graphical abstract
... The ErGPCR-1 mediates the 20HE signal through G-protein alpha q (Gαq) and phospholipase C gamma 1 (PLCG1), subsequently initiating Ca 2+ influx (Zhao et al., 2020). The intracellular Ca 2+ signaling triggers protein kinase C (PKC) to phosphorylate ultraspiracle (USP), an obligate partner for ecdysone receptor (EcR), and stimulates gene transcription (Tomaschko, 1999;Champlin and Truman, 2000;Srivastava et al., 2005;Losel and Wehling, 2003;Cai et al., 2014;Liu et al., 2014;Petruccelli et al., 2020;Zhao 2020). In addition, Ca 2+ signaling was involved in modulation of the YO responsiveness to MIH through Ca 2+ /calmodulin-dependent PDE as reported in P. clarkii (Nakatsuji et al., 2006;Mykles and Chang, 2020). ...
Article
Recently, genes in the superfamily of GPCR are gaining more interest in crustaceans as more evidence shows that they are involved in molting. This study identified four forms of the secretin family of G-protein coupled receptor (GPCR) from the Y-organ of mud crab, Scylla olivacea (ScoGPCR). A full-length sequence of ScoGPCR-B2 was isolated and identified as a lipoprotein receptor while three forms of GPCR in Methuselah-like (Mthl) or B3 subfamilies were reported as ScoGPCR-B3a, -B3b, and -B3c. These four forms exhibit common features of the 7-trans membrane (7TM) domain and distinct aspects in the extracellular region (ECR) at the N-terminus. At the ECR, disulfide bridges are predicted to generate structural stability in all four forms while the putative ScoGPCR-B3 proteins retain conserved Tyr, Trp, Pro, and Phe residues, possibly to form the aromatic-proline interactions and function as key residues for receptor recognition. Expression levels of ScoGPCR-B2 and -B3 in eyestalk, thoracic ganglion, and hindgut between intermolt and premolt stages are similar. Only ScoGPCR-B2 and ScoGPCR-B3a in Y-organ (YO) seem to be premolt-specific responses. An upregulation of ScoGPCR-B2 in YO at the premolt stage is correlated with the demand for cholesterol used in ecdysteroid synthesis, resulting in increased ecdysteroid titers. The effects of ecdysone on YO were pursued by in vitro incubation and revealed that ScoGPCR-B3a and -B3b expressions were induced in a different time frame: early in ScoGPCR-B3b and late in ScoGPCR-B3a. The early response of ScoGPCR-B3b was followed through immunohistology and showed that the newly synthesized protein was located primarily in the cytosol.
... The dopamine/ecdysteroid receptor (DopEcR) transmits the non-genomic signal of insect molting hormone 20hydroxyecdysone (20E) in Drosophila (Srivastava et al., 2005) and in Helicoverpa armigera (Kang et al., 2019). To date, several GPCRs have been proven to transmit 20E signals in H. armigera (Zhao, 2020), including ecdysone-responsible GPCR 1 (ErGPCR-1), ecdysone-responsible GPCR 2 (ErGPCR-2), and ecdysoneresponsible GPCR 3 (ErGPCR-3) (Cai et al., 2014a;Wang et al., 2015;Kang et al., 2021). These data suggest that several GPCRs function as steroid hormone receptors; however, whether any other GPCRs transmit 20E signals, and the mechanism by which several GPCRs function in 20E signaling, are unclear. ...
Article
Full-text available
G protein-coupled receptors (GPCRs) are the largest family of membrane receptors in animals and humans, which transmit various signals from the extracellular environment into cells. Studies have reported that several GPCRs transmit the same signal; however, the mechanism is unclear. In the present study, we identified all 122 classical GPCRs from the genome of Helicoverpa armigera , a lepidopteran pest species. Twenty-four GPCRs were identified as upregulated at the metamorphic stage by comparing the transcriptomes of the midgut at the metamorphic and feeding stages. Nine of them were confirmed to be upregulated at the metamorphic stage. RNA interference in larvae revealed the prolactin-releasing peptide receptor (PRRPR), smoothened (SMO), adipokinetic hormone receptor (AKHR), and 5-hydroxytryptamine receptor (HTR) are involved in steroid hormone 20-hydroxyecdysone (20E)-promoted pupation. Frizzled 7 (FZD7) is involved in growth, while tachykinin-like peptides receptor 86C (TKR86C) had no effect on growth and pupation. Via these GPCRs, 20E regulated the expression of different genes, respectively, including Pten (encoding phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase), FoxO (encoding forkhead box O), BrZ7 (encoding broad isoform Z7), Kr-h1 (encoding Krüppel homolog 1), Wnt (encoding Wingless/Integrated) and cMyc , with hormone receptor 3 (HHR3) as their common regulating target. PRRPR was identified as a new 20E cell membrane receptor using a binding assay. These data suggested that 20E, via different GPCRs, regulates different gene expression to integrate growth and development.
... In particular, molecular mechanisms of ecdysteroid secretion and cellular uptake have been revealed only recently in D. melanogaster larvae (Okamoto et al., 2018;Yamanaka et al., 2015), and further investigation in other life stages as well as in other insect species is clearly warranted. Recent studies have also elucidated non-canonical ecdysteroid signalling pathways, such as those mediated by G protein-coupled receptors (Cai et al., 2014;Ishimoto et al., 2013;Kang et al., 2019;Manaboon et al., 2009;Srivastava et al., 2005;Wang et al., 2015;Zhao, 2020). Such non-genomic actions of ecdysteroids can modulate the canonical signalling pathway mediated by EcR/USP, but they also have their own independent functions. ...
Chapter
Ecdysteroids are a group of steroid hormones originally identified as moulting hormones in insects. Ever since the isolation of the first ecdysteroid, studies in multiple model insect species have revealed their modes of action and pleiotropic functions. It is now established that they control a multitude of physiological processes, including moulting, metamorphosis, reproduction, and stress responses. Here I summarize our current understanding of the diverse functions of ecdysteroids throughout the insect life cycle. I then highlight six steps of ecdysteroid signalling as a framework to comprehend this complex hormone signalling pathway: biosynthesis, secretion, circulation, transport, modification, and gene expression regulation.
Article
Oviposition is induced upon mating in most insects. Spawning is a physiological process that is fundamental for the reproduction of Scylla paramamosain . However, the molecular mechanisms underlying the spawning process in this species are poorly understood. Herein, comprehensive ovary transcriptomic analysis was conducted at the germinal vesicle breakdown stage (GVBD), spawning stage, 0.5 h post‐spawning stage, and 24 h post‐spawning stage of S. paramamosain for gene discovery. A total of 67,230 unigenes were generated, and 27,975 (41.61%) unigenes were annotated. Meanwhile, the differentially expressed genes (DEGs) between the different groups were identified, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was subsequently conducted. These results suggested that octopamine (OA) and tyramine (TA) could induce oviposition, while dopamine (DA) and serotonin (5‐hydroxytryptamine [5‐HT]) inhibit oviposition. The 20‐hydroxyecdysone (20E) and methyl farnesoate (MF) signal pathways might be positively associated with oviposition. Furthermore, numerous transcripts that encode neuropeptides and their G‐protein‐coupled receptors (GPCRs), such as CNMamide, RYamide, ecdysis‐triggering hormone (ETH), GPA2/GPB5 receptor, and Moody receptor, appear to be differentially expressed during the spawning process. Eleven unigenes were selected for qRT‐PCR and the pattern was found to be consistent with the transcriptome expression pattern. Our work is the first spawning‐related investigation of S. paramamosain focusing on the ovary at the whole transcriptome level. These findings assist in improving our understanding of spawning regulation in S. paramamosain and provide information for oviposition studies in other crustaceans.
Article
Full-text available
Discontinuous muscle growth during molting is an important feature of Eriocheir sinensis. Molting is a physiological process completed by the cooperation of multiple organs. Signal transmission is critical for the accurate regulation of each step in molting. However, the knowledge of the signal transduction mechanism in the molting process of E. sinensis is presently very limited. In this work, the chromatin accessibility and gene expression of the muscle in E. sinensis in pre-molt (D) and post-molt (A) stages were sequenced by assay of transposase accessible chromatin sequencing (ATAC-seq) and RNA-seq, respectively. The differentially expressed genes (DEGs) in the muscle before and after molting were analyzed by combining ATAC-seq and RNA-seq, especially the G-protein coupled receptor (GPCR) genes in the process of signal transduction. The results showed that there were 616 common DEGs in ATAC-seq and RNA-seq in A vs. D stages, of which 538 were upregulated and 78 were downregulated. In the 19 DEGs included in the signaling transduction process, 13 were located in the GPCR signaling pathway and all were upregulated in A stages, which indicated that GPCRs play a leading role in muscle signal transmission during post-molt stage in molting. In these genes, the structure of the proteins encoded by 10 membrane-located genes with transmembrane activity was further analyzed. Six candidate GPCR genes were finally identified and further verified by real-time quantitative PCR (qRT-PCR). The GPCRs include metabotropic glutamate receptor 7, Mth-like 4, and Mth2 proteins. These results show the existence of GPCRs in the muscle of E. sinensis and, for the first time, found their dominant role in the signal transduction process during molting. It provides important clues for the study of muscle discontinuous growth and molting mechanism of E. sinensis.
Article
Full-text available
Polyphagous Apolygus lucorum has become the dominant insect in Bacillus thuringiensis (Bt) cotton fields. Hormone 20-hydroxyecdysone (20E) regulates multiple insect development and physiology events. 20E responses are controlled by pathways triggered by phospholipase C (PLC)-associated proteins. However, 20E-modulated genes and related proteins that can be affected by PLC still remain unknown. Here, isobaric tag for relative and absolute quantitation (iTRAQ) and immunoblotting techniques were used to compare differentially expressed proteins (DEPs) in A. lucorum in response to the treatment of 20E and the PLC inhibitor U73122 as well as their combination. A total of 1,624 non-redundant proteins and 97, 248, 266 DEPs were identified in the 20E/control, U73122/control, and 20E + U73122/control groups, respectively. Only 8 DEPs, including pathogenesis-related protein 5-like, cuticle protein 19.8, trans-sialidase, larval cuticle protein A2B-like, cathepsin L1, hemolymph juvenile hormone-binding protein, ATP-dependent RNA helicase p62-like, and myosin-9 isoform X1, were detected in all three groups. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the DEPs were involved in diverse signaling pathways. The results were validated by immunoblotting, which highlighted the reliability of proteomics analysis. These findings provided novel insights into the function of PLC in 20E signaling pathway in A. lucorum .
Article
In recent years, increasing numbers of microRNAs (miRNAs) have been reported to regulate insect metamorphosis. 1154 miRNAs have been previously identified from Tribolium castaneum by high-throughput sequencing; however, little is known about which miRNAs can participate in metamorphosis, leaving the role of miRNAs in regulating the underlying mechanism elusive. Here, we report the participation of miR-3017b in metamorphosis of T. castaneum. Temporal profiles revealed that miR-3017b was highly expressed at the late larval stage, but significantly decreased at the early pupal stage. Overexpression of miR-3017b caused larval to pupal to adult metamorphosis arrested. Dual luciferase reporter assay and miRNA–mRNA interaction assay illustrated that miR-3017b interacts with coding sequence of sarco/endoplasmic reticulum Ca²⁺ ATPase (SERCA) and suppresses its expression. Knockdown of SERCA caused metamorphosis arrested, similar to that observed in miR-3017b overexpression beetles. Further functional mechanism analyses revealed that 20-hydroxyecdysone application downregulates miR-3017b and up-regulates SERCA expression. The expression level of downstream genes in 20E pathway was disrupted after overexpressing miR-3017 and knockdown of SERCA. These results provided evidence miR-3017b-SERCA contributes to metamorphosis by regulating 20E pathway in T. castaneum. It could advance our understanding of the coordination of 20E and miRNA regulation in insect metamorphosis. This article is protected by copyright. All rights reserved.
Article
Insect herbivores use phytochemicals as signals to induce expression of their phytochemical-detoxifying cytochrome P450 monooxygenases (P450 s). The regulatory cascades that transduce phytochemical signals to enhanced expression of P450 s are the focus of this review. At least seven signaling pathways, including RTK/MAPK, GPCR/CREB, GPCR/NFκB, ROS/CncC/Keap1, AhR/ARNT, cytosol NR, and nucleus-located NR, may be involved in phytochemical induction of P450 s. Constitutive overexpression, over-phosphorylation, and/or activation of one or more effectors in the corresponding pathway are common causes of P450 overexpression that lead to phytochemical or insecticide resistance. Future research should pay more attentions to the starting point of each pathway, the number of pathways and their cross talk for a given phytochemical, and the pathways for downregulation of P450s.
Article
Proliferation of neural precursors in the optic lobe of Manduca sexta is controlled by circulating steroids and by local production of nitric oxide (NO), Diaphorase staining, anti-NO synthase (NOS) immunocytochemistry and the NO-indicator, DAF-2, show that cells throughout the optic anlage contain NOS and produce NO. Signaling via NO inhibits proliferation in the anlage, When exposed to low levels of ecdysteroid, NO production is stimulated and proliferation ceases, When steroid levels are increased, NO production begins to decrease within 15 minutes independent of RNA or protein synthesis and cells rapidly resume proliferation. Resumption of proliferation is not due simply to the removal of NO repression though, but also requires an ecdysteroid stimulatory pathway. The consequence of these opposing pathways is a sharpening of the responsiveness to the steroid, thereby facilitating a tight coordination between development of the different elements of the adult visual system.
Article
Heat shock protein 90 (Hsp90) interacts with steroid hormone receptors, signaling kinases, and various transcription factors. However, the mechanism by which Hsp90 interacts with different proteins in various pathways remains unclear. Western blot was used to study Hsp90 expression profile in Helicoverpa armigera (Lepidoptera). RNA interference was performed to investigate the function of Hsp90 in 20-hydroxyecdysone (20E) and juvenile hormone (JH) signal pathways. The binding of Hsp90 to the transcription factor ultraspiracle protein (USP1) and JH candidate receptor methoprene-tolerant (Met1) was analyzed by co-immunoprecipitation. Phospho-(Ser) PKC substrate antibody was used to detect Hsp90 phosphorylation. Hsp90 participated in 20E- or JH-induced gene expression. 20E induced the interaction between Hsp90 and USP1, whereas JH III and methoprene induced the interaction between Hsp90 and Met1, respectively. 20E and JH counteracted each other for these protein interactions. Both JH III and methoprene induced protein kinase C (PKC) phosphorylation of Hsp90. This process could be inhibited by phospholipase C (PLC) and PKC inhibitors. 20E suppressed JH III- or methoprene-induced PKC phosphorylation of Hsp90. 20E maintained the non-PKC-phosphorylation status of Hsp90. Hsp90 interacted with USP1 to induce gene expression in the 20E pathway. JH maintained the PKC-phosphorylation status of Hsp90. Hsp90 also interacted with Met1 to induce gene expression in the JH pathway. Our study describes a novel function of Hsp90 by altering phosphorylation and demonstrating that Hsp90 interacts with different proteins in various hormonal signaling pathways.
Article
Oestrogens are now recognised to be able to initiate rapid, fast responses, in addition to their classical, longer term actions. There is a growing appreciation of the potential implications of this mode of action for oestrogenic signalling in both neuronal and non-neuronal systems. As such there has been much effort to determine the mechanisms that are critical for transducing these rapid effects into cellular responses. Recently, an orphan G-protein coupled receptor (GPCR), termed GPR30, was identified as an oestrogen sensitive receptor in cancer cells. This receptor, now term G-protein estrogen receptor 1 (GPER1) has been the subject of many investigations, and a role for this receptor in the nervous system is now emerging. In this review, we wish to highlight some of the more recent advances in our understanding of the distribution and subcellular localisation of this receptor in the brain, as well as some of the evidence for the potential role that this receptor may play in the brain. We will then go on to discuss some of the controversies surrounding the pharmacology of this receptor, and attempt to reconcile them by suggesting that the "agonist-specific coupling" model of GPCR function may provide a potential explanation for some of the divergent reports of GPER1 pharmacology. This article is protected by copyright. All rights reserved.
Article
G protein-coupled receptors (or GPCRs) represent the largest family of membrane proteins in the human genome and are the target of approximately half of all therapeutic drugs. GPCRs contain a conserved structure of seven transmembrane domains. Their amino terminus is located extracellularly, whereas the carboxy terminus extends into the cytoplasm. Accumulating evidence suggests that GPCRs exist and function as monomeric entities. Nevertheless, more recent findings indicate that GPCRs can also form dimers or even higher order oligomers. The differential pharmacological and signaling properties of GPCR heteromeric complexes hint that their physiological effects may be different as compared to those obtained in tissue cultures that express a particular GPCR. In this chapter, we review current data on the role of GPCR heteromerization in receptor signaling, as well as its potential implication in neuropsychiatric disorders such as schizophrenia, depression, and Parkinson's disease.
Article
The activity of the glucocorticoid receptor (GR), a nuclear receptor transcription factor belonging to subclass 3C of the steroid/thyroid hormone receptor superfamily, is typically triggered by glucocorticoid hormones. Apart from driving gene transcription via binding onto glucocorticoid response elements in regulatory regions of particular target genes, GR can also inhibit gene expression via transrepression, a mechanism largely based on protein:protein interactions. Hereby GR can influence the activity of other transcription factors, without contacting DNA itself. GR is known to inhibit the activity of a growing list of immune-regulating transcription factors. Hence, GCs still rule the clinic for treatments of inflammatory disorders, notwithstanding concomitant deleterious side effects. Although patience is a virtue when it comes to deciphering the many mechanisms GR uses to influence various signaling pathways, the current review is testimony of the fact that groundbreaking mechanistic work has been accumulating over the past years and steadily continues to grow.
Article
Voltage-gated ion channels are transmembrane proteins that control nerve impulses and cell homeostasis. Signaling molecules that regulate ion channel activity and density at the plasma membrane must be specifically and efficiently coupled to these channels in order to control critical physiological functions such as action potential propagation. Although their regulation by G-protein receptor activation has been extensively explored, the assembly of ion channels into signaling complexes of GPCRs plays a fundamental role, engaging specific downstream -signaling pathways that trigger precise downstream effectors. Recent work has confirmed that GPCRs can intimately interact with ion channels and serve as -chaperone proteins that finely control their gating and trafficking in subcellular microdomains. This chapter aims to describe examples of GPCR-ion channel co-assembly, focusing mainly on signaling complexes between GPCRs and voltage-gated calcium channels.
Article
TRPV6, a highly calcium-selective member of the transient receptor potential (TRP) channel superfamily, is a major pathway for calcium absorption in the fetal and adult body. It is expressed abundantly in the duodenum, the placenta and exocrine tissues. TRVP6 was postulated to contribute to store-operated calcium channel (SOC) activity in certain cell types such as exocrine cells. In this study, we tested 2-APB, a widely used SOC inhibitor on human TRPV6 (hTRPV6) activity using fluorescence imaging, patch clamp and radioactive tracer techniques in transiently and stably transfected HEK293 cells. We found that the basal calcium and cadmium influx was higher in HEK293 cells transfected with hTRPV6 than in non-transfected cells. 2-APB inhibited hTRPV6 activity in both transient and stably transfected cells. This effect was slightly sensitive toward extracellular calcium. The extracellular sodium concentration did not affect the inhibition of hTRPV6 by 2-APB. However, N-methyl-d-glucamine significantly diminished the inhibitory effect of 2-APB presumably through direct interaction with this compound. Furthermore, 2-APB inhibited the activity of TRPV6 orthologs but not human TRPV5. 2-APB may serve as a parental compound for the development of therapeutic strategies specifically targeting the hTRPV6 calcium channel.
Article
The insect midgut undergoes programmed cell death (PCD) during metamorphosis, but the molecular basis for this phenomenon has not been demonstrated. We report a mod(mdg4) protein [designated as mod(mdg4)1A] that is involved in hormonally regulated insect midgut PCD, from the lepidopteran Helicoverpa armigera. Mod(mdg4)1A is localized in the larval midgut and is highly expressed during metamorphosis. Knockdown of mod(mdg4)1a by feeding dsRNA to the larvae suppressed midgut PCD and delayed metamorphosis. The mechanism is that mod(mdg4)1a knockdown decreased the transcript levels of genes involved in PCD and metamorphosis, but increased the transcript level of inhibitor of apoptosis survivin. The transcript level of mod(mdg4)1a is independently upregulated by 20-hydroxyecdysone (20E) or juvenile hormone (JH) analog methoprene. Overlapped 20E and methoprene counteractively regulate the transcript level of mod(mdg4)1a. 20E upregulates the mod(mdg4)1a transcript level not through its nuclear receptor EcRB1. Methoprene upregulates the mod(mdg4)1a transcript level through the juvenile hormone candidate receptor Met. These findings indicate that mod(mdg4)1a participates in midgut PCD and metamorphosis by regulating the transcript levels of a network of genes via different pathways under 20E and JH regulation.
Article
G protein coupled receptors (GPCRs) represent the largest family of cell surface receptors that are involved in regulating several physiological and behavioral responses in organisms. Indeed, over half of all the approved drugs on the market target GPCRs. Over the past twenty years, several lines of evidence have suggested that GPCRs associate to form oligomeric structures that substantially expand the complexity of signaling processes in vivo. In addition, GPCRs have also been shown to functionally regulate ion channels and help fine-tune neurotransmission. In this review, we will discuss recent advances in both mechanisms, with specific focus on opioid receptors, cannabinoid receptors and transient receptor potential (TRP) calcium channels in nociception. A better understanding of such mechanisms will be imperative in designing analgesics devoid of deleterious side effects and mitigating drug abuse.