You are on page 1of 25

JBC Papers in Press. Published on November 17, 2016 as Manuscript M116.

763235
The latest version is at http://www.jbc.org/cgi/doi/10.1074/jbc.M116.763235

G protein-coupled receptors (GPCRs) that signal via protein kinase A (PKA) cross-talk at insulin
receptor substrate 1 (IRS1) to activate the PI3K/AKT pathway

Nathan C. Lawa, Morris F. Whiteb, and Mary E. Hunzicker-Dunna,1

a
School of Molecular Biosciences, Washington State University, Pullman, WA 99164
b
Division of Endocrinology, Department of Medicine, Boston Children’s Hospital, Harvard Medical
School, Boston, Massachusetts 02115
1
Corresponding author: School of Molecular Biosciences, Washington State University, Biotechnology
Life Sciences Building Room 233, Pullman, WA 99164-7520. Phone: 509-335-5614, Email:

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


mehd@wsu.edu

Running title: GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

Keywords: G protein-coupled receptor (GPCR), protein kinase A (PKA), insulin-like growth factor 1
(IGF1), phosphatidylinositol-3 kinase (PI3K), insulin receptor substrate 1 (IRS1)

ABSTRACT and preovulatory granulosa cells, suggesting that


the PKA-dependent dephosphorylation of IRS1
GPCRs (G protein-coupled receptors)2 activate Ser/Thr residues is a conserved mechanism by
PI3K/AKT (phosphatidylinositol-3 kinase/v-AKT which GPCRs signal to activate the PI3K/AKT
thymoma viral oncoprotein) to regulate many pathway downstream of the IGF1R.
cellular functions that promote cell survival,
proliferation, and growth. However, the
mechanism by which GPCRs activate PI3K/AKT Cells respond to an array of stimuli
remains poorly understood. We used ovarian through both GPCRs (G protein-coupled
preantral granulosa cells (GCs) to elucidate the receptors) and RTKs (receptor tyrosine kinases)
mechanism by which the GPCR agonist FSH under normal physiological conditions.
(follicle-stimulating hormone) via PKA (protein Conversely, persistent activation of GPCRs or
kinase A) activates the PI3K/AKT cascade. IGF1 RTKs often drives mitogenic and metabolic
(insulin-like growth factor 1) is secreted in an responses that underlie tumorigenesis (1,2).
autocrine/paracrine manner by GCs and activates GPCRs and RTKs signal through the PI3K/AKT
the IGF1R (IGF1 receptor), but in the absence of (phosphatidylinositol-3 kinase/v-AKT thymoma
FSH fails to stimulate YXXM phosphorylation of viral oncoprotein) pathway to orchestrate a broad
IRS1 (insulin receptor substrate 1) required for spectrum of cellular functions (3-5). Due to its
PI3K/AKT activation. We show that PKA directly influential role in proliferation, apoptosis,
phosphorylates the PP1 (protein phosphatase 1) migration, and metabolism, the PI3K/AKT
regulatory subunit MYPT1 (myosin phosphatase pathway is actively pursued as a therapeutic target
targeting subunit 1) to activate PP1 associated to treat dysregulated glucose homeostasis, autism,
with the IGF1RŸIRS1 complex. Activated PP1 is infertility, cancer, and the diverse pathologies of
sufficient to dephosphorylate at least four IRS1 aging (6-9). However, the mechanism by which
Ser residues, Ser318, Ser346, Ser612, and Ser789, that GPCRs activate the PI3K/AKT pathway, either
promotes IRS1 YXXM phosphorylation by the alone or in conjunction with RTKs, is poorly
IGF1R and activates the PI3K/AKT cascade. understood (3,10).
Additional experiments indicate that this The RTKs for insulin and IGF1 (insulin-
mechanism also occurs in breast cancer, thyroid, like growth factor 1) phosphorylate YXXM (Tyr-

Copyright 2016 by The American Society for Biochemistry and Molecular Biology, Inc.
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

X-X-Met) motifs on the adapter proteins IRS1 (PKI) that functions as a catalytic subunit
(insulin receptor substrate 1) and IRS2 that bind to pseudosubstrate stimulated or blocked,
and activate class I PI3K p85-p110 heterodimers respectively, the phosphorylation of these markers
(11,12). PI3K generates the intracellular second of PI3K/AKT pathway activation (30).
messenger phosphatidylinositol-3,4,5-trisposphate FSH-stimulated PI3K/AKT activation is
that promotes phosphorylation of AKT on Thr308 also dependent on the IGF1R, based on the ability
by phosphoinositide-dependent kinase 1 (13). of the IGF1R antagonist NVP-AEW541 to block
AKT is also phosphorylated on Ser473 by the phosphorylation of IRS1(Tyr989), AKT(Thr308),
mechanistic target of rapamycin complex 2 (14). and AKT(Ser473) in response to FSH (31). The
Dual phosphorylation of AKT is necessary for full IGF1R is constitutively activated by secreted IGF1
activation (15); however, Thr308 is generally (~0.3 ng/ml) but the receptor fails to
considered a more reliable indicator of PI3K phosphorylate IRS1 YXXM motifs or activate
activity (7). AKT regulates many cellular PI3K/AKT in the absence of FSH (31).
functions through direct Ser/Thr phosphorylation PP1 is also required for FSH-stimulated

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


of key signaling intermediates as well as PI3K/AKT activation, evidenced by the ability of
transcription factors, such as FOXO1 (forkhead the PP1 inhibitor tautomycin as well as
box O1) (4). knockdown of the PP1 β catalytic subunit to
In contrast to the insulin and IGF1 significantly inhibit phosphorylation of
receptors, GPCR-mediated activation of PI3K is IRS1(Tyr989), AKT(Thr308), and AKT(Ser473) in
poorly understood (10). A number of GPCR response to FSH (31). However, the mechanism
ligands are known to activate PI3K, including by which FSH signals through PKA to stimulate
stromal cell-derived factor, sphingosine-1- IRS1 YXXM phosphorylation and PI3K/AKT
phosphate, lysophosphatidic acid, carbachol, activation in a PP1-dependent manner downstream
isoproterenol, prostaglandin E2, TSH (thyroid- of the IGF1R is not understood.
stimulating hormone), FSH (follicle-stimulating Blockade of the PI3K/AKT pathway by
hormone), LH/CG (luteinizing pharmacological inhibitors, a dominant-negative
hormone/choriogonadotropin) and others (10,16- AKT, or inactivation of the AKT target FOXO1
22). To date, the predominant mechanisms by abrogates the ability of FSH to promote GC
which GPCRs activate PI3K are often tissue- proliferation as well to induce genes that define
specific (10) and involve either the binding of the preovulatory (PO) GC such as the LH/CG
active Ras to the p110 catalytic subunit of PI3K receptor and aromatase (33-35). Thus, this
(23), an event that can be enhanced upon pathway and the genes it regulates are necessary
phosphorylation of the p85 regulatory subunit by for fertility (36-38).
PKA (protein kinase A) (19,24,25) or by Gβγ (26), In light of the physiological relevance of
or direct binding of Gβγ to p110 (17,26-29). the PI3K/AKT pathway, in this report we sought
We used ovarian preantral GCs (granulosa to investigate the mechanism by which PKA
cells) to model the mechanism by which the stimulates IRS1 YXXM phosphorylation and
GPCR agonist FSH activates the PI3K/AKT PI3K/AKT activation in a PP1-dependent manner
cascade. Our previous work showed that FSH- downstream of the IGF1R. We show that FSH via
stimulated PI3K/AKT activation is independent of PKA activates PP1, present in a complex that
Ras (30) but dependent on PKA, the IGF1R, and includes IGF1R and IRS1, by directly
PP1 (protein phosphatase 1) (30,31). phosphorylating one of its regulatory subunits to
The requirement for PKA in FSH- promote the dephosphorylation at least four IRS1
stimulated PI3K/AKT activation was Ser/Thr residues that facilitates IRS1 YXXM
demonstrated by results showing that a phosphorylation and PI3K/AKT activation.
constitutively active PKA catalytic mutant (PKA- Moreover, we show in three additional cell types
CQR) mimicked FSH to promote phosphorylation that the IGF1R-catalyzed phosphorylation of IRS1
of IRS1 on the YXXM motif Tyr989 and AKT and resulting PI3K activation is mediated by an
phosphorylation on Thr308 and Ser473 (31,32). This apparently conserved PKA- and PP1-dependent
result supports previous studies showing that mechanism that promotes the dephosphorylation
pharmacological activators or an inhibitor of PKA of IRS1 Ser/Thr residues.

2
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

RESULTS (myosin light chain) is a direct target of PP1 (51)


and served as a positive control for tautomycin
FSH stimulates multisite dephosphorylation of efficacy. GSK3β (glycogen synthase kinase 3β) is
Ser/Thr residues on IRS1 via PKA and PP1. a PP2 target (31) utilized to confirm the selectivity
Insulin resistance is associated with of tautomycin for PP1 over PP2 (52) (Figure 2B).
Ser/Thr phosphorylation of IRS1 that impairs
insulin-stimulated IRS1 YXXM phosphorylation PP1 and its regulatory subunit MYPT1 are
and PI3K/AKT activation (7,39). We used a panel associated with IRS1.
of monoclonal antibodies (40) against IRS1 Co-immunoprecipitation experiments
Ser/Thr phosphorylation sites (numbered from GC lysates showed that PP1cβ (the β isoform
according to rat amino acid sequence) and of the PP1 catalytic subunit) was constitutively
immunoblot analysis to evaluate IRS1 Ser/Thr associated with the complex of IRS1 and IGF1R
phosphorylation in response to FSH stimulation of (Figure 3A). We found that the PP1 regulatory
preantral GCs isolated from the ovaries of subunit MYPT1 was also constitutively associated

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


immature rats (31). While most IRS1 Ser/Thr with the IRS1ŸIGF1R complex (Figure 3A),
phosphorylation was not changed by FSH, FSH consistent with its role to facilitate substrate
significantly stimulated the phosphorylation specificity, catalytic regulation, and subcellular
Ser1216, Ser1135, Ser891, Ser857, Ser637, and Ser332/336 localization of PP1 catalytic subunits (51).
1.5-fold or greater (Figure 1). FSH also promoted Additional co-immunoprecipitation studies
the dephosphorylation of four Ser sites on IRS1 showed that PP1cβ and MYPT1 were
(Ser789, Ser612, Ser346, and Ser318) 1.5-fold or constitutively associated in GCs (Figures 3B and
greater. Importantly, Ser789, Ser612, and Ser318 are C). These results are consistent with our previous
previously reported as inhibitory sites on IRS1 studies that shRNA knockdown of PP1cβ blocks
(7,41-43). Unexpectedly, IRS1(Ser307) FSH-stimulated IRS1 YXXM phosphorylation and
phosphorylation that is usually detected during PI3K/AKT activation (31).
insulin resistance (7,44-47) was undetectable in
GCs. PKA activates PP1 through phosphorylation of
To evaluate the PKA and PP1 dependence MYPT1.
of IRS1 Ser/Thr dephosphorylation, cultured PKA phosphorylates MYPT1 on several
preantral GCs were pretreated with adenoviruses residues, although the regulatory effect and
expressing either GFP or the PKA-selective physiological significance of PKA-mediated
inhibitor PKI (protein kinase inhibitor) (48) MYPT1 phosphorylation remains unknown
(Figure 2A and C-E). Alternatively, GCs were (reviewed in (53)). We postulated that direct PKA
pretreated with lactacystin to block the phosphorylation of MYPT1 activates PP1cβ. To
degradation of IRS1 (31,49), and incubated test this hypothesis, the MYPT1ŸPP1cβ complex
without or with the PP1 inhibitor tautomycin (50) was isolated from GC lysates by
(Figure 2B and F-H). FSH-stimulated immunoprecipitation and treated under cell-free
dephosphorylation of Ser318, Ser346, and Ser612 was conditions with either H2O or recombinant PKA
inhibited by both PKI and tautomycin. FSH- catalytic protein (PKAc). Subsequent
stimulated dephosphorylation of Ser789 was also immunoblots probed for phospho-MYPT1(Ser668)
inhibited by PKI and tautomycin, as previously (Figure 4A) showed that PKAc phosphorylated
reported (31). FSH-stimulated dephosphorylation MYPT1; MYPT1(Ser668) is contained within the
of Ser612 in the absence of tautomycin was not sequence Arg-Arg-Arg-Ser668 that matches the
significant by ANOVA due to the larger and more PKA consensus phosphorylation motif (Arg-
variable signal from tautomycin-treated samples; Arg/Lys-X-Ser/Thr) (13). Similarly, immunoblot
however, Ser612 dephosphorylation was significant analysis of whole GC lysates showed that FSH
by Student’s t-test (p<0.05, Figure 2H). CREB stimulated the phosphorylation of MYPT1(Ser668)
(cAMP response element binding protein) is a in intact GCs, and this phosphorylation was
direct phosphorylation target of PKA in GCs (30) inhibited by the PKA-selective inhibitor (48) Myr-
served as a positive control for PKI treatments and PKI (myristoylated PKI) (Figure 4B-C).
a negative control for tautomycin treatments. MLC

3
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

Catalytic activity associated with the 5A, bound samples, compare lanes 2 versus 3).
MYPT1ŸPP1cβ complex was measured using the Probes for total MYPT1 indicated the presence of
protein phosphatase fluorogenic substrate immunoprecipitation-isolated MYPT1 (bound
DiFMUP (6,8-difluoro-4-methylumbelliferyl) samples, lanes 1 and 2), and longer exposures
(54,55). GCs were treated with vehicle or FSH for indicated endogenous MYPT1 isolated with the
15 min, and MYPT1 was isolated from lysates by IRS1ŸIGF1R complex (bound samples, lane 3).
immunoprecipitation. MYPT1 immunoprecipitates To test whether PP1cβŸMYPT1 was
were incubated without or with PKAc, and sufficient to dephosphorylate IRS1 and stimulate
phosphatase activity was measured via IRS1 YXXM phosphorylation in intact GCs, GCs
fluorescence emission from DiFMUP. PKAc were transfected with plasmids expressing GFP or
activated MYPT1 isolated from vehicle-treated a truncated (to residues 1-300) constitutively
GCs (Figure 4D). Interestingly, MYPT1 isolated active (CA) form of MYPT1 (56). Transfection of
from FSH-treated lysates had no significant GCs with CA MYPT1 stimulated
phosphatase activity, which might be attributed to dephosphorylation of IRS1(Ser318) in vehicle-

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


Ser668 dephosphorylation during treated cells (Figure 5B and E, compare lanes 1
immunoprecipitation isolation. However, the and 2). Additionally, expression of CA MYPT1 in
dephosphorylation of the PP1cβ direct target MLC GCs stimulated phosphorylation of IRS1(Tyr989), a
(51) from whole-cell lysates indicated FSH- canonical YXXM motif (Tyr989-X-X-Met), and
stimulated PP1cβ activity in primary cultures of AKT(Thr308) equivalent to that of FSH (Figure 5B-
GCs (Figure 2B). D, compare lanes 2 and 3). Probes for total
Finally, to assess phosphatase activity MYPT1 identified both endogenous MYPT1
bound to the IRS1ŸIGF1R complex, the complex (~140 kDa) and truncated CA MYPT1 (~30 kDa).
was isolated by immunoprecipitation using anti- Taken together, these results illustrated that
IGF1R antibody from vehicle-treated cells (as in PP1cβŸMYPT1 was necessary and sufficient to
Figure 3A) and incubated under cell-free dephosphorylate Ser sites on IRS1 and promote
conditions without or with PKAc. PKAc increased IRS1 YXXM phosphorylation and PI3K/AKT
the phosphatase activity associated with the activation.
IRS1ŸIGF1R complex by approximately 2.5-fold
(Figure 4E). GPCR-mediated activation of the PI3K/AKT
pathway downstream of the IGF1R is a
PP1cβ/MYPT1 promotes IRS1 Ser conserved mechanism among different cell
dephosphorylation and IRS1 YXXM types.
phosphorylation. Based on the widespread expression of
Next, we investigated whether PKAc- IGF1R in mammals (2,57), we hypothesized that
activated PP1cβŸMYPT1 dephosphorylated our model for GPCR-mediated activation of the
Ser/Thr residues on IRS1. PP1cβŸMYPT1 was PI3K/AKT pathway downstream the IGF1R in
isolated by immunoprecipitation using an preantral GCs might be conserved among different
antibody against MYPT1 and treated without cell types. To test this hypothesis, we analyzed the
(designated “MYPT1 IP”) or with PKAc PKA- and IGF1R-dependence of markers for
(designated “Activated MYPT1 IP”) (as in PI3K/AKT activation in response to elevated
Figures 4A and D). ‘MYPT1 IP’ or ‘Activated cAMP in primary, serum-free cultures of rat PO
MYPT1 IP’ samples were combined with GCs as well as serum-starved cultures of MCF7
IRS1ŸIGF1R isolated by immunoprecipitation (as human breast cancer and FRTL rat thyroid cell
in Figure 3A) and incubated at room temperature lines (for culture conditions refer to “Experimental
in phosphatase assay buffer. The combination of Procedures”). In response to FSH, preantral GCs
PKAc-activated PP1cβŸMYPT1 and IRS1ŸIGF1R (used in the experiments above) differentiate to
isolated from vehicle-treated cells was sufficient PO GCs that are phenotypically distinct from
to dephosphorylate IRS1(Ser318) (Figure 5A, preantral GCs (20).
bound samples, compare lanes 1 versus 2) Cells were pretreated with the IGF1R-
comparable to that detected in IRS1ŸIGF1R selective inhibitor NVP-AEW541 (58) or the
immunoprecipitates of FSH-treated GCs (Figure PKA-selective inhibitor PKI (either Myr-PKI or

4
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

adenoviral PKI). MCF7 and FRTL cells were then GCs to elucidate how GPCRs can activate the
treated with the adenylyl cyclase activator PI3K/AKT pathway downstream of the IGF1R.
forskolin to elevate intracellular levels of cAMP, Several studies report that the GPCR
while PO GCs were treated with hCG (human agonist FSH stimulates activation of the
chorionic gonadotropin), a GPCR agonist for the PI3K/AKT pathway in preantral GCs (31-
LH/CG receptor that signals predominately via 33,35,60,61). In a previous study, we reported that
cAMP and PKA (reviewed in ref 20). GCs secrete low levels of IGF1 that activate the
Human-CG in PO GCs and forskolin in IGF1R, but fail to stimulate IRS1 YXXM
MCF7 and FRTL cells significantly stimulated phosphorylation in the absence of FSH (31).
IRS1(Tyr989) and AKT(Thr308) phosphorylation as Furthermore, both PKA and PP1 are necessary for
well as the concomitant phosphorylation of FSH-stimulated IRS1 YXXM phosphorylation and
MYPT1(Ser668) and dephosphorylation of PI3K/AKT activation (31). Here, we show that
IRS1(Ser318) over vehicle controls (Figure 6 and PKA activates PP1 via direct phosphorylation of
Supplemental Figure S1). Pretreatment with NVP- the PP1 regulatory subunit MYPT1. PP1 and

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


AEW541 abolished hCG- or forskolin-stimulated MYPT1 associate with the IRS1ŸIGF1R complex
IRS1(Tyr989) and AKT(Thr308) phosphorylation and mediate the dephosphorylation of four Ser
(Figure 6 A-B, G-H, and M-N). Additionally, residues on IRS1, three of which are previously
pretreatment with PKI blocked hCG- or forskolin- described as inhibitory to PI3K activation (7,41-
stimulated IRS1(Ser318) dephosphorylation as well 43). Importantly, activated PP1cβŸMYPT1 mimics
as MYPT(Ser668), IRS1(Tyr989), and AKT(Thr308) FSH-stimulated IRS1 Ser dephosphorylation,
phosphorylation (Figure 6 C-F, I-L, and O-R). It is IRS1 YXXM phosphorylation, and PI3K/AKT
worth noting that in MCF7 cells treated with Myr- activation. Therefore, our results support the
PKI, forskolin-stimulated MYPT(Ser668) hypothesis (depicted in Figure 7) that FSH signals
phosphorylation was statistically different from via PKA to activate PP1 and promote IGF1R
vehicle controls (p<0.05), but significantly signaling via dephosphorylation of IRS1 Ser
inhibited (63.1±5.6% compared to H2O controls, residues to facilitate IRS1 YXXM phosphorylation
p<0.05; Figure 6 O). Also, basal phosphorylation that activates the PI3K/AKT cascade.
of IRS1(Ser318) was decreased in the presence of Phospho-proteomic analysis of L6 skeletal
Myr-PKI in MCF7 cells (Figure 6P), a result muscle cells previously revealed the interaction of
which might reflect secondary effects on pathways MYPT1 and IRS1 (62) as well as the potential role
that stimulate basal IRS1(Ser318) phosphorylation. of MYPT1 in insulin responses (63); however, the
Blockade of forskolin- and hCG-stimulated role of MYPT1 was unknown. We show that CA
activation of the PI3K pathway by PKI established MYPT1 (in the absence of FSH) stimulates IRS1
that forskolin and hCG promoted the cAMP- Ser dephosphorylation, IRS1 YXXM
dependent activation of PKA in MCF7, FRTL, and phosphorylation, and PI3K/AKT activation.
PO GCs. Together, these data illustrated that Additionally, no significant additive affect was
cAMP-stimulated activation of the PI3K/AKT seen when CA MYPT1-expressing GCs were
pathway required both the IGF1R and PKA in rat treated with FSH (see Figure 5B-E, compare lanes
PO GCs, human MCF7 breast cancer cells, and rat 3 and 4), suggesting that the ability of FSH to
FTRL thyroid cells. stimulate IRS1 YXXM phosphorylation and PI3K
activation is predominantly mediated by the
DISCUSSION activation of PP1cβŸMYPT1 and the
dephosphorylation of IRS1 Ser/Thr residues.
GPCRs constitute the largest family of The inhibitory potential of IRS1 Ser/Thr
cell-surface proteins that transduce extracellular phosphorylation is supported by decades of
stimuli through complex signaling networks research on insulin signaling (reviewed in
(1,59). Among the signaling cascades regulated by (7,39,64)). Several studies report that mutation of
GPCRs, the PI3K/AKT pathway intersects several select IRS1 Ser/Thr residues to Ala enhances
vital cell functions, including apoptosis, insulin-stimulated IRS1 YXXM phosphorylation
proliferation, metabolism, growth, and glucose and PI3K/AKT activation (recently reviewed in
homeostasis (3-5). We utilized ovarian preantral (7)). Additionally, Ser/Thr phosphorylation of

5
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

IRS1 by various kinases is correlated with pathway. Based on the near-ubiquitous expression
diminished insulin-stimulated PI3K activation of the IGF1R and PKA (73,74), understanding the
(40). However, not all IRS1 Ser/Thr mechanism by which PKA can intersect
phosphorylation is inhibitory. For example, downstream of the IGF1R will likely shed new
mutation of IRS1(Ser1216) (human residue Ser1223) light on how GPCRs impact normal and aberrant
to Ala decreases insulin-stimulated IRS1 YXXM physiological responses. For example, activating
phosphorylation in CHO cells expressing the mutations that induce tumorigenesis occur in a
insulin receptor (65). Interestingly, FSH number of GPCRs that activate PKA, including
stimulated the phosphorylation of IRS1(Ser1216) in the receptors for TSH, FSH, LH, neuromedin B,
GCs, suggesting that activating phosphorylation(s) gastrin, and others (1). Therefore, in the context
might contribute to FSH-stimulated PI3K/AKT of cancer, the potential to activate PI3K/AKT
activation. We previously reported that PKA through mutated GPCRs may account for the
promotes the direct phosphorylation of IRS1, altered metabolism, inhibited apoptosis, and
although the contribution of this phosphorylation enhanced proliferation associated with

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


to PI3K/AKT activation was not directly evaluated tumorigenesis. Thus, the molecular link between
(31). Further studies are necessary to understand GPCRs and the PI3K/AKT pathway is a valuable
the integrated regulatory role of multisite IRS1 contribution to the ongoing development of
phosphorylation in various tissue backgrounds. targeted therapeutic approaches to treating a broad
FSH regulates broad shifts in gene range of cancers and other pathologies (9,75).
expression that drive the maturation of ovarian Clearly, further studies are warranted to evaluate
preantral follicles to a preovulatory phenotype both the extent to which our proposed model is
capable of responding to the surge of LH that conserved among PKA-dependent systems and its
triggers ovulation and the resumption of meiosis in therapeutic potential.
oocytes ((34) and reviewed in (20)). The use of
dominant negative or constitutively active AKT EXPERIMENTAL PROCEDURES
mutants or PI3K inhibitors illustrate that the
PI3K/AKT pathway plays a pivotal role in Materials – Materials were previously described
coordinating many vital gene expression responses (31) with the following additions: antibody
to FSH in preantral GCs (33,35,66-70). FOXO recognizing total MYPT1 (for immunoblot and IP)
family transcription factors are the predominant was purchased from BD Biosciences; pregnant
transcriptional regulators downstream of AKT that mare’s serum gonadotropin (PMSG) and forskolin
function as either activators or repressors (4,71). were purchased from Sigma-Aldrich Co., LLC;
In GCs, FOXO1 regulates approximately 60% of hCG was purchased from Abraxis Pharmaceutical
over 3,700 genes up- or down-regulated by FSH Products; Enzchek® Phosphatase Assay kit
(34). Additionally, inactivation of the genes containing DiFMUP, OptiMEM, and
encoding FOXO1 and FOXO3 in murine GCs Lipofectamine® 2000 were purchased from
impairs follicle maturation and fertility (72). Thus, Thermo Fisher Scientific, Inc. The following
transcriptional responses downstream of the antibodies were purchased from Cell Signaling
PI3K/AKT pathway are critical for normal Technology (primary dilution indicated): GAPDH
maturation of ovarian follicles and hence, fertility. (1:1000), phospho-IRS1(Ser318) (1:1000 in 5%
GPCRs activate the PI3K/AKT pathway BSA + 0.1% Tween), phospho-IRS1(Ser332/336)
in a number of tissues across many mammals, (1:500 in 5% BSA + 0.1% Tween), phospho-
including FSH receptors in preantral GCs, LH/CG IRS1(Ser612) (1:750), phospho-IRS1(Ser635)
1100
receptors in PO GCs, β-adrenergic receptors in (1:500), phospho-IRS1(Ser ) (1:1000), and
human MCF7 breast cancer cells, and TSH phospho-MYPT1(Ser668) (1:333).
receptors in rat thyroid cells (19,20,25). Each of
these ligand-activated GPCRs can signal via PKA Granulosa, FRTL, and MCF7 Cell Culture –
(19,25,32). Our results support a conserved Preantral GCs were isolated from 24-day-old
mechanism beyond preantral ovarian GCs by immature Sprague-Dawley rats primed with
which GPCRs/PKA cross-talk downstream of the estrogen and cultured under serum-free conditions
IGF1R at IRS1 to activate the PI3K/AKT as previously described (31). PO GCs were

6
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

isolated from 24-day-old immature rats injected on ice. Phosphatase activity was then assayed at
with PMSG 48 h prior to isolation and cultured room temperature from 455 nm fluorescence
under serum-free conditions (76). Rats were from emission (360 nm excitation) measured over time
a breeder colony (originally from Charles River on a Victor X5 Multilabel plate reader
Laboratory) maintained in accordance with the (PerkinElmer, Inc.). Accumulation of fluorescence
Animal Welfare Act by protocols approved by within the linear range was used to calculate
Washington State University Animal Care and Use phosphatase activity.
Committee. Transduction of preantral and PO GCs
with adenoviruses followed procedures used Cell-free Dephosphorylation of IRS1 – MYPT1
previously (67). and IRS1 were isolated by immunoprecipitation
FRTL cells were obtained from American using antibodies against MYPT1 and IGF1R,
Type Culture Collection (ATCC), and MCF7 respectively. Immunoprecipitations were
luminal A subtype, estrogen receptor positive cells separately collected in the absence of NaF and
were provided by Dr. Ruth Keri (Case Western Na2P2O as in phosphatase assay procedure above.

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


Reserve University). FRTL and MCF7 cells were Washed MYPT1 pull-down samples were treated
subcultured no more than three times to avoid with H2O or recombinant PKA catalytic subunit
changes due to passaging and were cultured according to previous protocols (31). MYPT1 and
according to protocols established by ATCC, IGF1R immunoprecipitations were then
except insulin was omitted from MCF7 cell resuspended in 1X phosphatase assay buffer,
complete medium. FRTL cells were plated at combined, and incubated on a rotator at room
~2x106 cells per well in 3 mL medium and MCF7 temperature for 3 h. Pellets were analyzed by
cells at ~1x106 in 3 mL followed by overnight immunoblot.
incubation. Serum-containing medium over FRTL
and MCF7 cells was then removed, replaced with Transfection of Preantral GCs – Prior to plating,
serum- and insulin-free medium, and cells 2.5x106 isolated preantral GCs were resuspended
incubated overnight before treatments were in 2 mL OptiMEM. Lipofectamine® 2000 and
conducted and sample collection. Notably, FRTL cDNA plasmids were combined according to the
and MCF7 cells were treated with Myr-PKI as manufacturer’s protocol, added to the resuspended
opposed to adenoviral PKI because overnight GCs, and the combined solution was immediately
transduction was lethal (data not shown). plated on fibronectin-coated plates. GCs were
incubated for 3 days in
Immunoblot, Immunoprecipitation, and cell-free OptiMEM/Lipofectamine®/cDNA solution before
PKA Phosphorylation Assay – Following medium was removed and replaced with
treatments, cells were collected and processed DMEM/F12 medium containing estrogen,
according to previously described procedures penicillin, and streptomycin (3131). GCs were
(31,77). incubated for >6 h, treated as indicated, and
collected for immunoblot analysis.
Phosphatase Assay – GCs were treated as
indicated and collected in a previously-described Statistical Analysis – Densitometry from
(31) immunoprecipitation lysis buffer without the immunoblots was determined using Quantity One
Ser/Thr phosphatase inhibitors NaF and Na2P2O7. 1-D Analysis Software (Bio-Rad Laboratories,
Pull-down (“bound”) samples were washed three Inc.) taken from images within the approximate
times as previously (31) and resuspended in 100 linear range of detection. Densitometry signal was
µL 1X phosphatase assay buffer containing 50 normalized to loading controls. Experiments were
mM Tris-HCl (pH 7.5) and 150 mM NaCl. analyzed by one-way ANOVA followed by
Resuspended immunoprecipitates were loaded into Tukey’s multiple comparisons test (p<0.05) or
a black 96-well clear-bottom plate (Corning, Inc.) Student’s t-test (p<0.05) using Prism software
and kept on ice. DiFMUP was diluted in 1X in (GraphPad Software, Inc.). Each experimental
phosphatase assay buffer according to the replicate (n) represents an independent experiment
manufacturer’s protocol and combined with performed on GCs collected from ovaries of ~ 8
immunoprecipitation samples in the 96-well plate

7
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

rats. Therefore, each n represents a biological replicate.

Acknowledgements: We thank Dr. Masumi Eto for the constitutively activate MYPT1 construct. We
also thank Dr. Ruth Keri for kindly providing MCF7 cells. This research was supported by National
Institute of Health (NIH) Grants R01HD065859 (to M.H.D.), R01HD062053 (to M.H.D.),
T32GM083864 (to N.C.L), R01DK098655-01 (to M.F.W.), and R01DK038712-22 (to M.F.W.).
Additional funding was provided by The Fund for Science (to M.H.D.) and the Poncin Scholarship Fund
(to N.C.L).

Conflict of Interest: The authors have no conflicts of interest for this article.

Author Contributions: N.C.L. designed and performed experiments and analyzed data with oversight
from M.E.H-D.; N.C.L., M.F.W., and M.E.H-D wrote the paper.

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


REFERENCES

1. Marinissen, M. J., and Gutkind, J. S. (2001) G-protein-coupled receptors and signaling


networks: emerging paradigms. Trends in pharmacological sciences 22, 368-376
2. Riedemann, J., and Macaulay, V. M. (2006) IGF1R signalling and its inhibition.
Endocrine-related cancer 13 Suppl 1, S33-43
3. Rozengurt, E., Sinnett-Smith, J., and Kisfalvi, K. (2010) Crosstalk between
insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling
systems: a novel target for the antidiabetic drug metformin in pancreatic cancer. Clinical
cancer research : an official journal of the American Association for Cancer Research
16, 2505-2511
4. Manning, B. D., and Cantley, L. C. (2007) AKT/PKB signaling: navigating downstream.
Cell 129, 1261-1274
5. Medina, D. L., and Santisteban, P. (2000) Thyrotropin-dependent proliferation of in vitro
rat thyroid cell systems. European journal of endocrinology / European Federation of
Endocrine Societies 143, 161-178
6. Hsueh, A. J., Kawamura, K., Cheng, Y., and Fauser, B. C. (2015) Intraovarian control of
early folliculogenesis. Endocrine reviews 36, 1-24
7. Copps, K. D., and White, M. F. (2012) Regulation of insulin sensitivity by
serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2.
Diabetologia 55, 2565-2582
8. Vivanco, I., and Sawyers, C. L. (2002) The phosphatidylinositol 3-Kinase AKT pathway
in human cancer. Nature reviews. Cancer 2, 489-501
9. Klempner, S. J., Myers, A. P., and Cantley, L. C. (2013) What a tangled web we weave:
emerging resistance mechanisms to inhibition of the phosphoinositide 3-kinase pathway.
Cancer discovery 3, 1345-1354
10. Vanhaesebroeck, B., Guillermet-Guibert, J., Graupera, M., and Bilanges, B. (2010) The
emerging mechanisms of isoform-specific PI3K signalling. Nature reviews. Molecular cell
biology 11, 329-341
11. Rordorf-Nikolic, T., Van Horn, D. J., Chen, D., White, M. F., and Backer, J. M. (1995)
Regulation of phosphatidylinositol 3'-kinase by tyrosyl phosphoproteins. Full activation
requires occupancy of both SH2 domains in the 85-kDa regulatory subunit. The Journal
of biological chemistry 270, 3662-3666
12. Shoelson, S. E., Chatterjee, S., Chaudhuri, M., and White, M. F. (1992) YMXM motifs of
IRS-1 define substrate specificity of the insulin receptor kinase. Proceedings of the
National Academy of Sciences of the United States of America 89, 2027-2031

8
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

13. Pearce, L. R., Komander, D., and Alessi, D. R. (2010) The nuts and bolts of AGC protein
kinases. Nature reviews. Molecular cell biology 11, 9-22
14. Yang, G., Murashige, D. S., Humphrey, S. J., and James, D. E. (2015) A Positive
Feedback Loop between Akt and mTORC2 via SIN1 Phosphorylation. Cell reports 12,
937-943
15. Vanhaesebroeck, B., and Alessi, D. R. (2000) The PI3K-PDK1 connection: more than
just a road to PKB. The Biochemical journal 346 Pt 3, 561-576
16. Guillermet-Guibert, J., Bjorklof, K., Salpekar, A., Gonella, C., Ramadani, F., Bilancio, A.,
Meek, S., Smith, A. J., Okkenhaug, K., and Vanhaesebroeck, B. (2008) The p110beta
isoform of phosphoinositide 3-kinase signals downstream of G protein-coupled receptors
and is functionally redundant with p110gamma. Proceedings of the National Academy of
Sciences of the United States of America 105, 8292-8297
17. Murga, C., Fukuhara, S., and Gutkind, J. S. (2000) A novel role for phosphatidylinositol
3-kinase beta in signaling from G protein-coupled receptors to Akt. The Journal of

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


biological chemistry 275, 12069-12073
18. Alvarez, C. J., Lodeiro, M., Theodoropoulou, M., Camina, J. P., Casanueva, F. F., and
Pazos, Y. (2009) Obestatin stimulates Akt signalling in gastric cancer cells through beta-
arrestin-mediated epidermal growth factor receptor transactivation. Endocrine-related
cancer 16, 599-611
19. Ciullo, I., Diez-Roux, G., Di Domenico, M., Migliaccio, A., and Avvedimento, E. V. (2001)
cAMP signaling selectively influences Ras effectors pathways. Oncogene 20, 1186-1192
20. Hunzicker-Dunn, M. E., and Mayo, K. E. (2015) Gonadotropin Signaling in the Ovary. in
Physiology of Reproduction (Zeleznik, A. J., and Plant, T. eds.), 4 Ed., Elsevier, San
Diego, CA, USA. pp 895-945
21. K1, M., H, I., K, M., M, T., S, F., H, K., H, A., Y, K., N, H., and Y, N. (2005) Activation of
PI3K-Akt pathway mediates antiapoptotic effects of beta-adrenergic agonist in airway
eosinophils. American journal of physiology. Lung cellular and molecular physiology
255(5), L860-867
22. HS, K., MY, J., MA, B., DD, M., and AM, G. (2011) Prostaglandin E2 activates and
utilizes mTORC2 as a central signaling locus for the regulation of mast cell chemotaxis
and mediator release. The Journal of biological chemistry 286, 391-402
23. Rodriguez-Viciana, P., Warne, P. H., Dhand, R., Vanhaesebroeck, B., Gout, I., Fry, M.
J., Waterfield, M. D., and Downward, J. (1994) Phosphatidylinositol-3-OH kinase as a
direct target of Ras. Nature 370, 527-532
24. Torella, D., Gasparri, C., Ellison, G. M., Curcio, A., Leone, A., Vicinanza, C., Galuppo,
V., Mendicino, I., Sacco, W., Aquila, I., Surace, F. C., Luposella, M., Stillo, G., Agosti, V.,
Cosentino, C., Avvedimento, E. V., and Indolfi, C. (2009) Differential regulation of
vascular smooth muscle and endothelial cell proliferation in vitro and in vivo by
cAMP/PKA-activated p85alphaPI3K. American journal of physiology. Heart and
circulatory physiology 297, H2015-2025
25. De Gregorio, G., Coppa, A., Cosentino, C., Ucci, S., Messina, S., Nicolussi, A., D'Inzeo,
S., Di Pardo, A., Avvedimento, E. V., and Porcellini, A. (2007) The p85 regulatory
subunit of PI3K mediates TSH-cAMP-PKA growth and survival signals. Oncogene 26,
2039-2047
26. Luttrell, L. M., van Biesen, T., Hawes, B. E., Koch, W. J., Touhara, K., and Lefkowitz, R.
J. (1995) G beta gamma subunits mediate mitogen-activated protein kinase activation by
the tyrosine kinase insulin-like growth factor 1 receptor. The Journal of biological
chemistry 270, 16495-16498
27. Stoyanov, B., Volinia, S., Hanck, T., Rubio, I., Loubtchenkov, M., Malek, D., Stoyanova,
S., Vanhaesebroeck, B., Dhand, R., Nurnberg, B., and et al. (1995) Cloning and

9
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

characterization of a G protein-activated human phosphoinositide-3 kinase. Science 269,


690-693
28. Kurosu, H., Maehama, T., Okada, T., Yamamoto, T., Hoshino, S., Fukui, Y., Ui, M.,
Hazeki, O., and Katada, T. (1997) Heterodimeric phosphoinositide 3-kinase consisting of
p85 and p110beta is synergistically activated by the betagamma subunits of G proteins
and phosphotyrosyl peptide. The Journal of biological chemistry 272, 24252-24256
29. Stephens, L., Smrcka, A., Cooke, F. T., Jackson, T. R., Sternweis, P. C., and Hawkins,
P. T. (1994) A novel phosphoinositide 3 kinase activity in myeloid-derived cells is
activated by G protein beta gamma subunits. Cell 77, 83-93
30. Hunzicker-Dunn, M. E., Lopez-Biladeau, B., Law, N. C., Fiedler, S. E., Carr, D. W., and
Maizels, E. T. (2012) PKA and GAB2 play central roles in the FSH signaling pathway to
PI3K and AKT in ovarian granulosa cells. Proceedings of the National Academy of
Sciences of the United States of America 109, E2979-2988
31. Law, N. C., and Hunzicker-Dunn, M. E. (2016) Insulin Receptor Substrate 1, the Hub

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


Linking Follicle-stimulating Hormone to Phosphatidylinositol 3-Kinase Activation. The
Journal of biological chemistry 291, 4547-4560
32. Puri, P., Little-Ihrig, L., Chandran, U., Law, N. C., Hunzicker-Dunn, M., and Zeleznik, A.
J. (2016) Protein Kinase A: A Master Kinase of Granulosa Cell Differentiation. Sci Rep 6,
28132
33. Zeleznik, A. J., Saxena, D., and Little-Ihrig, L. (2003) Protein kinase B is obligatory for
follicle-stimulating hormone-induced granulosa cell differentiation. Endocrinology 144,
3985-3994
34. Herndon, M. K., Law, N. C., Donaubauer, E. M., Kyriss, B., and Hunzicker-Dunn, M.
(2016) Forkhead box O member FOXO1 regulates the majority of follicle-stimulating
hormone responsive genes in ovarian granulosa cells. Molecular and cellular
endocrinology 434, 116-126
35. Park, Y., Maizels, E. T., Feiger, Z. J., Alam, H., Peters, C. A., Woodruff, T. K., Unterman,
T. G., Lee, E. J., Jameson, J. L., and Hunzicker-Dunn, M. (2005) Induction of cyclin D2
in rat granulosa cells requires FSH-dependent relief from FOXO1 repression coupled
with positive signals from Smad. The Journal of biological chemistry 280, 9135-9148
36. Sicinski, P., Donaher, J. L., Geng, Y., Parker, S. B., Gardner, H., Park, M. Y., Robker, R.
L., Richards, J. S., McGinnis, L. K., Biggers, J. D., Eppig, J. J., Bronson, R. T., Elledge,
S. J., and Weinberg, R. A. (1996) Cyclin D2 is an FSH-responsive gene involved in
gonadal cell proliferation and oncogenesis. Nature 384, 470-474
37. Lei, Z. M., Mishra, S., Zou, W., Xu, B., Foltz, M., Li, X., and Rao, C. V. (2001) Targeted
disruption of luteinizing hormone/human chorionic gonadotropin receptor gene.
Molecular endocrinology 15, 184-200
38. KL, B., AE, D., VA, C., M, D., NG, W., ME, J., ER, S., and JK, F. (2000) An age-related
ovarian phenotype in mice with targeted disruption of the Cyp 19 (aromatase) gene.
Endocrinology 141, 2614-2623
39. Sun, X. J., and Liu, F. (2009) Phosphorylation of IRS proteins Yin-Yang regulation of
insulin signaling. Vitamins and hormones 80, 351-387
40. Hancer, N. J., Qiu, W., Cherella, C., Li, Y., Copps, K. D., and White, M. F. (2014) Insulin
and metabolic stress stimulate multisite serine/threonine phosphorylation of insulin
receptor substrate 1 and inhibit tyrosine phosphorylation. The Journal of biological
chemistry 289, 12467-12484
41. Tzatsos, A., and Tsichlis, P. N. (2007) Energy depletion inhibits phosphatidylinositol 3-
kinase/Akt signaling and induces apoptosis via AMP-activated protein kinase-dependent
phosphorylation of IRS-1 at Ser-794. The Journal of biological chemistry 282, 18069-
18082

10
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

42. Mothe, I., and Van Obberghen, E. (1996) Phosphorylation of insulin receptor substrate-1
on multiple serine residues, 612, 632, 662, and 731, modulates insulin action. The
Journal of biological chemistry 271, 11222-11227
43. Weigert, C., Kron, M., Kalbacher, H., Pohl, A. K., Runge, H., Haring, H. U., Schleicher,
E., and Lehmann, R. (2008) Interplay and effects of temporal changes in the
phosphorylation state of serine-302, -307, and -318 of insulin receptor substrate-1 on
insulin action in skeletal muscle cells. Molecular endocrinology 22, 2729-2740
44. Copps, K. D., Hancer, N. J., Opare-Ado, L., Qiu, W., Walsh, C., and White, M. F. (2010)
Irs1 serine 307 promotes insulin sensitivity in mice. Cell metabolism 11, 84-92
45. Herrema, H., Lee, J., Zhou, Y., Copps, K. D., White, M. F., and Ozcan, U. (2014)
IRS1Ser(3)(0)(7) phosphorylation does not mediate mTORC1-induced insulin
resistance. Biochemical and biophysical research communications 443, 689-693
46. Aguirre, V., Werner, E. D., Giraud, J., Lee, Y. H., Shoelson, S. E., and White, M. F.
(2002) Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


the insulin receptor and inhibits insulin action. The Journal of biological chemistry 277,
1531-1537
47. Rui, L., Aguirre, V., Kim, J. K., Shulman, G. I., Lee, A., Corbould, A., Dunaif, A., and
White, M. F. (2001) Insulin/IGF-1 and TNF-alpha stimulate phosphorylation of IRS-1 at
inhibitory Ser307 via distinct pathways. The Journal of clinical investigation 107, 181-189
48. Cheng, H. C., Kemp, B. E., Pearson, R. B., Smith, A. J., Misconi, L., Van Patten, S. M.,
and Walsh, D. A. (1986) A potent synthetic peptide inhibitor of the cAMP-dependent
protein kinase. The Journal of biological chemistry 261, 989-992
49. Lee, A. V., Gooch, J. L., Oesterreich, S., Guler, R. L., and Yee, D. (2000) Insulin-like
growth factor I-induced degradation of insulin receptor substrate 1 is mediated by the
26S proteasome and blocked by phosphatidylinositol 3'-kinase inhibition. Molecular and
cellular biology 20, 1489-1496
50. Swingle, M., Ni, L., and Honkanen, R. E. (2007) Small-molecule inhibitors of ser/thr
protein phosphatases: specificity, use and common forms of abuse. Methods in
molecular biology 365, 23-38
51. Bollen, M., Peti, W., Ragusa, M. J., and Beullens, M. (2010) The extended PP1 toolkit:
designed to create specificity. Trends in biochemical sciences 35, 450-458
52. Chen, X., Zheng, Y., and Shen, Y. (2007) Natural products with maleic anhydride
structure: nonadrides, tautomycin, chaetomellic anhydride, and other compounds.
Chemical reviews 107, 1777-1830
53. Butler, T., Paul, J., Europe-Finner, N., Smith, R., and Chan, E. C. (2013) Role of serine-
threonine phosphoprotein phosphatases in smooth muscle contractility. American journal
of physiology. Cell physiology 304, C485-504
54. Ni, L., Swingle, M. S., Bourgeois, A. C., and Honkanen, R. E. (2007) High yield
expression of serine/threonine protein phosphatase type 5, and a fluorescent assay
suitable for use in the detection of catalytic inhibitors. Assay and drug development
technologies 5, 645-653
55. Fontal, O. I., Vieytes, M. R., Baptista de Sousa, J. M., Louzao, M. C., and Botana, L. M.
(1999) A fluorescent microplate assay for microcystin-LR. Analytical biochemistry 269,
289-296
56. Eto, M., Kirkbride, J. A., and Brautigan, D. L. (2005) Assembly of MYPT1 with protein
phosphatase-1 in fibroblasts redirects localization and reorganizes the actin
cytoskeleton. Cell motility and the cytoskeleton 62, 100-109
57. Arcaro, A. (2013) Targeting the insulin-like growth factor-1 receptor in human cancer.
Frontiers in pharmacology 4, 30
58. Garcia-Echeverria, C., Pearson, M. A., Marti, A., Meyer, T., Mestan, J., Zimmermann, J.,
Gao, J., Brueggen, J., Capraro, H. G., Cozens, R., Evans, D. B., Fabbro, D., Furet, P.,

11
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

Porta, D. G., Liebetanz, J., Martiny-Baron, G., Ruetz, S., and Hofmann, F. (2004) In vivo
antitumor activity of NVP-AEW541-A novel, potent, and selective inhibitor of the IGF-IR
kinase. Cancer cell 5, 231-239
59. Kroeze, W. K., Sheffler, D. J., and Roth, B. L. (2003) G-protein-coupled receptors at a
glance. Journal of cell science 116, 4867-4869
60. Baumgarten, S. C., Convissar, S. M., Fierro, M. A., Winston, N. J., Scoccia, B., and
Stocco, C. (2014) IGF1R signaling is necessary for FSH-induced activation of AKT and
differentiation of human Cumulus granulosa cells. The Journal of clinical endocrinology
and metabolism 99, 2995-3004
61. Kinoshita, E., Kinoshita-Kikuta, E., and Koike, T. (2012) Phos-tag SDS-PAGE systems
for phosphorylation profiling of proteins with a wide range of molecular masses under
neutral pH conditions. Proteomics 12, 192-202
62. Geetha, T., Langlais, P., Luo, M., Mapes, R., Lefort, N., Chen, S. C., Mandarino, L. J.,
and Yi, Z. (2011) Label-free proteomic identification of endogenous, insulin-stimulated

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


interaction partners of insulin receptor substrate-1. Journal of the American Society for
Mass Spectrometry 22, 457-466
63. Zhang, X., Ma, D., Caruso, M., Lewis, M., Qi, Y., and Yi, Z. (2014) Quantitative
phosphoproteomics reveals novel phosphorylation events in insulin signaling regulated
by protein phosphatase 1 regulatory subunit 12A. Journal of proteomics 109, 63-75
64. White, M. F. (2002) IRS proteins and the common path to diabetes. American journal of
physiology. Endocrinology and metabolism 283, E413-422
65. Luo, M., Reyna, S., Wang, L., Yi, Z., Carroll, C., Dong, L. Q., Langlais, P., Weintraub, S.
T., and Mandarino, L. J. (2005) Identification of insulin receptor substrate 1
serine/threonine phosphorylation sites using mass spectrometry analysis: regulatory role
of serine 1223. Endocrinology 146, 4410-4416
66. Liu, Z., Rudd, M. D., Hernandez-Gonzalez, I., Gonzalez-Robayna, I., Fan, H. Y.,
Zeleznik, A. J., and Richards, J. S. (2009) FSH and FOXO1 regulate genes in the
sterol/steroid and lipid biosynthetic pathways in granulosa cells. Molecular endocrinology
23, 649-661
67. Law, N. C., Weck, J., Kyriss, B., Nilson, J. H., and Hunzicker-Dunn, M. (2013) Lhcgr
expression in granulosa cells: roles for PKA-phosphorylated beta-catenin, TCF3, and
FOXO1. Molecular endocrinology 27, 1295-1310
68. Li, Q., He, H., Zhang, Y. L., Li, X. M., Guo, X., Huo, R., Bi, Y., Li, J., Fan, H. Y., and Sha,
J. (2013) Phosphoinositide 3-kinase p110delta mediates estrogen- and FSH-stimulated
ovarian follicle growth. Molecular endocrinology 27, 1468-1482
69. Wang, X. L., Wu, Y., Tan, L. B., Tian, Z., Liu, J. H., Zhu, D. S., and Zeng, S. M. (2012)
Follicle-stimulating hormone regulates pro-apoptotic protein Bcl-2-interacting mediator of
cell death-extra long (BimEL)-induced porcine granulosa cell apoptosis. The Journal of
biological chemistry 287, 10166-10177
70. Mani, A. M., Fenwick, M. A., Cheng, Z., Sharma, M. K., Singh, D., and Wathes, D. C.
(2010) IGF1 induces up-regulation of steroidogenic and apoptotic regulatory genes via
activation of phosphatidylinositol-dependent kinase/AKT in bovine granulosa cells.
Reproduction 139, 139-151
71. Van Der Heide, L. P., Hoekman, M. F., and Smidt, M. P. (2004) The ins and outs of
FoxO shuttling: mechanisms of FoxO translocation and transcriptional regulation. The
Biochemical journal 380, 297-309
72. Liu, Z., Ren, Y. A., Pangas, S. A., Adams, J., Zhou, W., Castrillon, D. H., Wilhelm, D.,
and Richards, J. S. (2015) FOXO1/3 and PTEN Depletion in Granulosa Cells Promotes
Ovarian Granulosa Cell Tumor Development. Molecular endocrinology 29, 1006-1024

12
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

73. Hartog, H., Wesseling, J., Boezen, H. M., and van der Graaf, W. T. (2007) The insulin-
like growth factor 1 receptor in cancer: old focus, new future. European journal of cancer
43, 1895-1904
74. Kirschner, L. S., Yin, Z., Jones, G. N., and Mahoney, E. (2009) Mouse models of altered
protein kinase A signaling. Endocrine-related cancer 16, 773-793
75. Yuan, T. L., and Cantley, L. C. (2008) PI3K pathway alterations in cancer: variations on
a theme. Oncogene 27, 5497-5510
76. Flynn, M. P., Fiedler, S. E., Karlsson, A. B., Carr, D. W., Maizels, E. T., and Hunzicker-
Dunn, M. (2016) Dephosphorylation of MAP2D enhances its binding to vimentin in
preovulatory ovarian granulosa cells. Journal of cell science 129, 2983-2996
77. Donaubauer, E. M., Law, N. C., and Hunzicker-Dunn, M. E. (2016) Follicle-Stimulating
Hormone (FSH)-dependent Regulation of Extracellular Regulated Kinase (ERK)
Phosphorylation by the Mitogen-activated Protein (MAP) Kinase Phosphatase MKP3.
The Journal of biological chemistry 291, 19701-19712

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


2
Abbreviations used: GPCR, G protein-coupled receptor; FSH, follicle-stimulating hormone; IGF1,
insulin-like growth factor 1; IRS1, insulin receptor substrate 1; GC, granulosa cell; IGF1R, insulin-like
growth factor 1 receptor; PI3K, phosphatidylinositol-3-kinase; AKT, v-AKT thymoma viral oncoprotein;
PKA, protein kinase A; PP1, protein phosphatase 1; CREB, cAMP response element-binding protein;
MLC, myosin light chain;

FIGURE LEGENDS

FIGURE 1. FSH stimulates the phosphorylation and dephosphorylation of several Ser/Thr residues
in IRS1. Primary cultures of rat preantral GCs were treated with vehicle (v) or FSH (F) for 15 min. Total
cell lysates were collected for immunoblot analysis using a panel of monoclonal antibodies that recognize
phosphorylated Ser/Thr residues within IRS1. A, Representative images are shown. Arrows indicate
bands of interest. B, Densitometric quantification of phosphorylations was normalized to load controls,
and fold FSH-stimulated phosphorylation is represented by Log10 scale. Black and green asterisks indicate
IRS1 residues significantly (p<0.05) phosphorylated or dephosphorylated 1.5-fold or greater with FSH,
respectively. Results are represented as the mean ± S.E. (error bars) of fold FSH-stimulate
phosphorylation (n = 3).

FIGURE 2. FSH-stimulated dephosphorylation of IRS1 Ser318, Ser346, and Ser612 are PKA- and PP1-
dependent. A and C-E, Preantral GCs were pretreated/transduced with adenoviruses expressing GFP (Ad-
GFP) or the PKA inhibitor PKI (Ad-PKI), treated with vehicle or FSH for 15 min, and collected for
immunoblot analysis, as described in legend to Figure 1. B and F-H, GCs were pretreated with 10 µM
lactacystin and ethanol (EtOH) or 1 µM tautomycin (Taut) for 5.5 h, followed by vehicle or FSH for 15
min, and collected. Asterisks indicate significantly (*, p<0.05; **, p<0.01; ****, p<0.0001) or not
significantly (n.s.) different signal compared to vehicle controls of the same pretreatment as analyzed by
one-way ANOVA and Tukey’s multiple comparisons test (n = 3). # indicates significantly (p<0.05)
different signal compared to vehicle controls of the same pretreatment as analyzed by Student’s t-test
(n=3). All data represent mean ± S.E. (error bars). Dotted lines indicate cropped image from the same
exposure.

FIGURE 3. PP1cβ and MYPT1 are constitutively associated with the complex of IRS1 and the
IGF1R. A, IRS1 and the IGF1R were isolated by immunoprecipitation (IP) from GC lysates treated with

13
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

vehicle or FSH using an antibody recognizing the IGF1R versus IgG controls. B-C, PP1cβ or MYPT1
were isolated by immunoprecipitation from GC lysates versus IgG controls. Each image from A-C is
representative of three independent experiments. Dotted lines separate different exposures of the same
probe. Input fractions represent ~6% of total protein and immunoprecipitates (“bound”) represent the
remaining fraction from which each target protein was isolated.

FIGURE 4. PKA phosphorylation of MYPT1 stimulates MYPT1-associated phosphatase activity. A,


MYPT1 was isolated by immunoprecipitation from untreated GC lysates. Immunoprecipitates were
incubated under cell-free conditions with H2O or recombinant PKA catalytic subunit (C), and subsequent
samples analyzed by immunoblot. Image represents three independent experiments. B-C, GCs were
pretreated with H2O or 50 µM myristoylated PKI (Myr-PKI), followed by vehicle or FSH for 15 min, and
whole-cell lysates analyzed by immunoblot. Asterisk indicates significant (*, p<0.05) or not significant
(n.s.) FSH-stimulated MYPT1(Ser668) phosphorylation signal as compared to vehicle controls of the same
pretreatment. Data represented as mean signal ± S.E. (error bars) analyzed by one-way ANOVA and

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


Tukey’s multiple comparison’s test (n = 3). D, MYPT1 was isolated by immunoprecipitation from GC
lysates treated with vehicle or FSH, treated with H2O (veh and FSH) or recombinant PKA catalytic
subunit (PKAc) (as in A), and phosphatase activity measured by accumulation of fluorescence over time
from DiFMUP (refer to “Experimental Procedures”). Data represent mean arbitrary fluorescent units per
min ± S.E. (error bars) analyzed by one-way ANOVA and Tukey’s multiple comparisons test (n = 3;
****, p<0.0001). E, IRS1 and IGF1R were isolated by immunoprecipitation using an antibody
recognizing the IGF1R, treated with H2O or PKAc (as in A), and phosphatase activity measured as in D.
Data represent mean arbitrary fluorescent units per min ± S.E. (error bars) analyzed by Student’s t-test (n
= 3; *, p<0.05).

FIGURE 5. MYPT1 is sufficient to stimulate IRS1 Ser/Thr dephosphorylation, IRS1 YXXM


phosphorylation, and PI3K/AKT activation. A, The IRS1ŸIGF1R complex was isolated by
immunoprecipitation (as in Figure 3A) from lysates of GCs treated with vehicle or FSH. Separately,
MYPT1 was isolated by immunoprecipitation and treated with H2O (MYPT1 IP) or PKAc (“Activated”
MYPT1 IP) (as in Figures 4A and D). Isolated IRS1ŸIGF1R complexes from vehicle-treated lysates were
combined with either MYPT1 IP (“bound” sample, lane 1) or “Activated” MYPT1 IP (“bound” sample,
lane 2) and incubated at room temperature for 3 h in phosphatase assay buffer. Results are representative
of two independent experiments. B-E, GCs were transfected with plasmids expressing either GFP or a
constitutively activate MYPT1 mutant (CA MYPT1), treated with vehicle or FSH, and total cell lysates
collected for immunoblot analysis. B, Representative images are shown. Dotted lines indicate cropped
image from the same exposure. C-E, Densitometric quantifications of target phosphorylations relative to
load controls are represented as mean signal ± S.E. (error bars). Samples with significantly different (*,
p<0.05, **, p<0.01) or not significantly different (n.s.) signal from other treatment groups are indicated
based on analysis by one-way ANOVA and Tukey’s multiple comparisons test (n = 3).

FIGURE 6. Inhibition of the IGF1R or PKA blocks PI3K/AKT pathway activation in response to
cAMP-producing compounds in rat preovulatory GCs, FRTL rat thyroid cells, and human MCF7
breast cancer cells. Primary cultures of rat preovulatory (PO) GCs (A-B) as well as FRTL (G-H) and
MCF7 (M-N) cell lines were pretreated with DMSO or 1 µM NVP-AEW541 (AEW) for 1 h. In separate
experiments, PO GCs (C-F) were transduced/pretreated with adenoviruses expressing either GFP (Ad-
GFP) or PKI (Ad-PKI), and FRTL (I-L) and MCF7 (O-R) cells were pretreated with H2O or 40 µM Myr-
PKI for 1 h. PO GCs were then treated with vehicle or 1 I.U./mL hCG for 15 min, and FRTL and MCF7
cells were treated with vehicle or 10 µM forskolin for 15 min. Total cell extracts were collected and
analyzed by immunoblot. Densitometric quantifications of target phosphorylations over load controls are
represented as mean signal ± S.E. (error bars). Samples with significantly different (*, p<0.05; **,
p<0.01; ***, p<0.001; ****, p<0.0001) or not significantly different (n.s.) signal compared to vehicle

14
GPCRs and IGF1R cross-talk at IRS1 to activate PI3K

controls of the same pretreatment are indicated based on analysis by one-way ANOVA and Tukey’s
multiple comparisons test (n = 3).

FIGURE 7. Proposed model of the mechanism by which GPCRs signal via PKA and PP1 to promote
PI3K/AKT pathway activation. Endogenous IGF1 secreted from cells activates the IGF1R, but fails to
stimulate IRS1 YXXM phosphorylation that drives PI3K activation due to the presence of Ser/Thr
phosphorylations on IRS1. PKA activated downstream GPCRs phosphorylates the PP1 regulatory subunit
MYPT1 to activate PP1. Activated PP1 then dephosphorylates Ser/Thr residues on IRS1, thereby
stimulating IGF1R-dependent IRS1 YXXM phosphorylation to activate the PI3K/AKT pathway. The
GPCR agonists FSH or hCG, the adenylyl cyclase activator forskolin, the PKA inhibitor PKI, a
constitutively activate (CA) MYPT1 mutant, the PP1 inhibitor tautomycin, and the IGF1R inhibitor NVP-
AEW541 were utilized in the experiments outlined herein.

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016

15
FIGURE S1. Pretreatment of rat preovulatory GCs, FRTL rat thyroid cell, and MCF7 human
breast cancer cells with IGF1R or PKA inhibitors blocks PI3K/AKT pathways activation in
response to cAMP-producing compounds. Primary cultures of rat preovulatory (PO) GCs (A) as well as
FRTL (B) and MCF7 (C) cell lines were pretreated with DMSO or 1 µM NVP-AEW541 for 1 h. In
separate experiments, PO GCs (D) were transduced/pretreated with adenoviruses expressing either GFP
(Ad-GFP) or PKI (Ad-PKI), and FRTL (E) and MCF7 (F) cells were pretreated with H2O or 40 µM Myr-
PKI for 1 h. PO GCs were then treated with vehicle or 1 I.U./mL hCG for 15 min, and FRTL and MCF7
cells were treated with vehicle or 10 µM forskolin for 15 min. Total cell extracts were collected and
analyzed by immunoblot. Representative images from three independent experiments are shown and
densitometric quantifications are located in Figure 6.
G protein-coupled receptors (GPCRs) that signal via protein kinase A (PKA)
cross-talk at insulin receptor substrate 1 (IRS1) to activate the PI3K/AKT pathway
Nathan C. Law, Morris F. White and Mary E. Hunzicker-Dunn
J. Biol. Chem. published online November 17, 2016

Access the most updated version of this article at doi: 10.1074/jbc.M116.763235

Alerts:
• When this article is cited
• When a correction for this article is posted

Downloaded from http://www.jbc.org/ at UNIV OF WEST FLORIDA on November 20, 2016


Click here to choose from all of JBC's e-mail alerts

Supplemental material:
http://www.jbc.org/content/suppl/2016/11/17/M116.763235.DC1.html

This article cites 0 references, 0 of which can be accessed free at


http://www.jbc.org/content/early/2016/11/17/jbc.M116.763235.full.html#ref-list-1

You might also like