You are on page 1of 9

ARTICLE IN PRESS

Ann Anat 187 (2005) 529—537

www.elsevier.de/aanat

Unique expression and regulatory mechanisms


of EG-VEGF/prokineticin-1 and its receptors
in the corpus luteum$
Tatiana Kisliouk, Helena Podlovni, Rina Meidan

Department of Animal Sciences, Faculty of Agricultural, Food and Environmental Quality Sciences, The Hebrew
University of Jerusalem, Rehovot 76100, Israel

Received 8 June 2005; accepted 1 July 2005

KEYWORDS Summary
Endothelial cells;
Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) or Prokineticin-
Luteal steroidogenic
1 (PK-1) is a novel cysteine-rich protein that belongs to the AVIT protein family. EG-
cells;
VEGF/PK-1, described as selective angiogenic mitogen, is widely expressed in
VEGF;
different tissues including steroidogenic endocrine glands. This review summarizes
Angiogenesis;
the expression and functions of EG-VEGF/PK-1 in corpus luteum (CL)-derived cells:
Apoptosis;
endothelial and steroidogenic cell types. EG-VEGF/PK-1 mRNA is expressed by luteal
Hypoxia
steroidogenic cells of human, rat and bovine ovaries, but was absent from the luteal
Endothelial cells CLEC. Luteal EC expressed high levels of both PK-receptors PK-R1 and
PK-R2 – the two G protein-coupled PK-1 receptors. Interestingly, expression of EG-
VEGF/PK-1 and VEGF were inversely regulated in human and bovine luteinized
granulosa cells. EG-VEGF/PK-1 elevated [3H]-thymidine incorporation, MAPK activa-
tion and c-jun/fos mRNA expression and enhanced LEC proliferation. EG-VEGF/PK-1
also inhibited serum starvation-induced apoptosis in these cells. Stress conditions such
as serum withdrawal, TNFa and chemical hypoxia markedly increase PK-R2 expression,
whereas mRNA levels of PK-R1 remain unchanged, implying that the anti-apoptotic
effect of PK-1 on LEC may be mediated via PK-R2. Besides its direct mitogenic and
anti-apoptotic effects, EG-VEGF/PK-1 elevated VEGF mRNA expression in bovine
luteal steroidogenic cells, which possesses only PK-R1. Together, these findings suggest
an important role for PK-1 in luteal function by acting as a mitogen and survival factor
in LEC. Nevertheless, the inverse regulation of EG-VEGF/PK1 and VEGF mRNA
expression by ovarian cells and the distribution of its receptors may suggest that in
addition to its angiogenic effects, EG-VEGF/PK-1 may also play other roles in ovary.
& 2005 Elsevier GmbH. All rights reserved.

$
Main Lecture for the 100th Meeting of the Anatomische Gesellschaft in Leipzig, Germany, 11–14 March 2005.
Corresponding author. Tel.: +972 89489394; fax: +972 89465763.
E-mail address: rina.meidan@huji.ac.il (R. Meidan).

0940-9602/$ - see front matter & 2005 Elsevier GmbH. All rights reserved.
doi:10.1016/j.aanat.2005.07.005
ARTICLE IN PRESS
530 T. Kisliouk et al.

Introduction Blood vessel formation also involves other essen-


tial factors, including the angiopoietins (Hazzard et
The corpus luteum (CL) is a short-lived endocrine al., 1999; Wulff et al., 2000; Fraser and Lunn,
gland which develops from the granulosa and theca 2001). Angiopoietin-1 (Ang-1) is a secreted growth
cells of the preovulatory follicle. The main role of factor which binds the Tie-2 receptor tyrosine
the CL is the production of progesterone, which is kinase. This factor enhances the stability of the
required for the establishment and maintenance of newly formed blood vessels (Wulff et al., 2000;
pregnancy by acting on the genital tract, most Fraser and Lunn, 2001). Angiopoietin-2 (Ang-2)
importantly on the uterus (Smith et al., 1994; destabilizes capillary integrity, thus facilitating
Niswender et al., 2000). sprouting by VEGF (Fraser and Lunn, 2001). At the
In the absence of embryonic signals the CL late luteal stage or during luteolysis mRNA levels of
undergoes luteolysis and eventually the tissue is Ang-1 sharply decrease, whereas mRNA levels of
eliminated by apoptosis (for review see McCracken Ang-2 increase (Fraser and Lunn, 2001; Wulff et al.,
et al., 1999). The development, function and 2001; Tanaka et al., 2004).
regression of the CL are controlled by a large Recently, LeCouter et al. reported the identifica-
number of regulators. In most mammals, luteinizing tion of a new angiogenic mitogen that promotes
hormone (LH) is the main luteotropin and prosta- proliferation, migration, fenestration and survival
glandin F2a (PGF2a) is the main luteolytic agent of adrenal cortical capillary endothelial cells (ACE)
(McCracken et al., 1999; Carambula et al., 2003; (LeCouter et al., 2001). As it was found in
Wang et al., 2003). Insulin-like growth factors (IGF-I endocrine glands and shown to act selectively on
and -II), growth hormone (GH), prolactin and EC derived from these glands, it was designated
steroid hormones can synergize with LH to regulate endocrine gland-derived vascular endothelial
ovarian function and were all shown to have direct growth factor (EG-VEGF; LeCouter et al., 2001).
effects on ovarian cells in different species (Zetser This review summarizes the expression and roles of
et al., 2001; Schams and Berisha, 2002, 2004; EG-VEGF in ovarian tissue with special emphasis on
Schams et al., 2002; Webb et al., 2002). the biological functions of this peptide in steroido-
The rapid cyclical changes in luteal growth and genic and EC of the CL.
regression are associated with rapid changes in
vasculature. Luteal development is accompanied
by a dramatic increase in the number of blood
vessels (Augustin et al., 1995; Fraser and Lunn, Prokineticin protein family: Structure
2001; Redmer et al., 2001). In the mid-stage and pattern of expression
of CL the vascular network is so extensive that
most steroidogenic cells appear to be juxtaposed EG-VEGF is identical to prokineticin 1 (PK-1)
to capillary vessels (Zheng et al., 1993). Endo- which has first been identified as a stimulant of
thelial cells (EC) of the CL account for approxi- gastrointestinal smooth muscle contraction (Li et
mately half of all cell types at mid-cycle (Young al., 2001). PK-1 is highly related to mamba
et al., 2000). Therefore, the CL provides a uni- intestinal toxin-1 (Li et al., 2001). These proteins
que model system for the study of the cellular together with the frog skin peptide Bv8 (Bombina
and molecular regulation of physiological angio- variegata) and its mammalian homolog prokineticin
genesis. VEGF is the main angiogenic factor 2 (PK-2) belong to the same protein family (Kaser et
in CL. Inhibition of VEGF (or its signaling) during al., 2003). PKs share 60% amino acid identity and a
the early or mid-luteal phase results in a marked common protein structural motif (Fig. 1). They are
reduction in luteal angiogenesis and suppression of small secreted proteins consisting of 80–90 amino
luteal function (Fraser and Wulff, 2003). High acids, including 10 conserved cysteines. Their
mRNA levels of VEGF and its type 2 receptor amino-termini are identical; therefore the se-
VEGFR-2 or KDR are found during early to quence of the first four residues, AVIT, was
mid-luteal phase and decrease at late luteal suggested as a name for this family of proteins
stage (Berisha et al., 2000; Sugino et al., 2001; (Kaser et al., 2003). The PKs are the cognate
Al-zi’abi et al., 2003; Fraser et al., 2005). The LH ligands for two highly homologous G protein-
surge augments VEGF production by granulosa cells coupled receptors, PK receptors (PK-R1 and PK-
of the ovulatory follicle (Hazzard et al., 1999). In R2) that are about 80% identical to a previously
addition to LH, hypoxia and IGFs further augment described mouse orphan receptor gpr73 (Lin et al.,
VEGF expression in these cells (Martinez-Chequer 2002a; Masuda et al., 2002; Soga et al., 2002).
et al., 2003). Its essential role has been demon- To the present date human, mouse, rat and
strated. bovine EG-VEGF/PK-1 have been cloned (LeCouter
ARTICLE IN PRESS
Role of prokineticin-1 and its corpus luteum 531

PK-2b MRSSRCARLLLLLLLPPLLLTPPAGDAAVITGACDRDPQCGGGMCCAVSLWVKSIRICTP 6
PK-2a MRSSRCARLLLLLLLPPLLLTPPAGDAAVITGACDRDPQCGGGMCCAVSLWVKSIRICTP 60
PK-1 ---------------------------AVITGACERDVQCRAGTCCAVSLWLRGLRVCTP 33
*******:** ** .* *******::.:*:***

PK-2b MGKVGDSCHPMTRKNHFGNGRQERRKRKRRRKKKVPFLGRRMHHTCPCLPGLACSRTSFN 120


PK-2a MGKVGDSCHPMTRK--------------------VPFLGRRMHHTCPCLPGLACSRTSFN 100
PK-1 LGRAGEECHPGSHK--------------------VPFFRKRQHHACPCLPNLLCSRGLDG 73
:*:.*:.*** ::* ***: :* **:*****.* *** .

PK-2b RYTCLAQK----- 128


PK-2a RYTCLAQK----- 108
PK-1 RYRCSTNLKNINF 86
** * ::

Figure 1. A comparison of amino-acid sequences of bovine prokineticins, PK-1 and PK-2 (a and b forms), respectively.
Signs used are as defined in CLUSTAL software W: an asterisk denotes identity in amino-acid residues; colons denote
conserved substitutions; full stops denote semi-conserved substitutions.

et al., 2001, 2003a; Masuda et al., 2002; Kisliouk et circadian rhythm (Cheng et al., 2002). High levels
al., 2005). The genomic structures of human and of PK-2 were demonstrated in the testis where it
mouse sequences have been conserved. Exon 1 was confined to primary spermatocytes (Wechsel-
contains the coding sequences for signal peptide berger et al., 1999; LeCouter et al., 2003b). PK-2 is
and the first 5 amino acids of the mature protein. encoded by four exons with the third exon
Exon 2 encodes 6 of 10 cysteins and 42 residues in subjected to alternative splicing. Exon 3 codes for
total, and exon 3 the remaining 49 amino acids 21 amino acids, of which 11 are lysines or arginins.
(LeCouter et al., 2003a). In human and rat EG- Therefore, the long form of PK-2 was termed basic
VEGF/PK1 mRNA expression has been described in a PK-2 (PK-2b). The shorter variant, lacking 21 amino
variety of tissues, in steroidogenic glands (such as acids, was consequently termed acidic PK-2 (PK-2a,
ovary, testis, and adrenal gland), but also in the Fig. 1) (Wechselberger et al., 1999; Kaser et al.,
gastrointestinal tract, nervous system, bladder, and 2003). Recently a shorter form of basic (or long) PK-
prostate (LeCouter et al., 2001, 2003b; Li et al., 2, PK-2b, was described: PK-2b is produced by
2001; Masuda et al., 2002). Several studies have proteolytic cleavage of PK-2b and in contrast to the
examined the expression pattern of EG- EG-VEGF/ parental molecule it selectively activates PK-R1
PK-1 in the human and primate ovaries; it was (Chen et al., 2005).
present in ovarian stroma, follicles and corpora Unlike the testis, PK-2 mRNA was almost un-
lutea (LeCouter et al., 2001; Ferrara et al., 2003; detectable in human and bovine ovaries (Ferrara et
Kisliouk et al., 2003; Fraser et al., 2005). Similarly al., 2003; Fraser et al., 2005; Kisliouk et al., 2005).
in the bovine species, luteal steroidogenic cells
derived from mid-cycle CL as well as follicular
theca and granulosa cells expressed EG-VEGF/PK-1
mRNA. However EG-VEGF/PK-1 mRNA was not Regulation of EG-VEGF/PK-1 expression
detected in luteal EC (Kisliouk et al., 2005). It in the ovarian steroidogenic cell types
was low at early-stage human CL (Ferrara et al.,
2003), but up-regulated during mid-luteal to late Luteinized human granulosa cells, those har-
luteal phase (Ferrara et al., 2003; Fraser et al., vested from patients undergoing IVF and SV-40
2005). Rather surprisingly, maximal expression of transformed granulosa cells (SVOG), as well as
EG-VEGF was found at very late luteal phase, at a bovine luteal steroidogenic cells express EG-VEGF/
time when VEGF expression is greatly reduced or PK-1 mRNA (Kisliouk et al., 2003). SVOG cells, like
undetectable (Fraser et al., 2005). non-transformed granulosa cells also produce VEGF,
The expression of PK-2 mRNA is somewhat more interestingly however, the expression of these two
restricted than PK-1, PK-2 was found in suprachias- pro-angiogenic factors was inversely regulated in
matic nucleus of the brain where it affects these cells. cAMP elevating agent, forskolin, was
ARTICLE IN PRESS
532 T. Kisliouk et al.

shown to be a potent stimulator of EG-VEGF/PK-1 elevating VEGF mRNA expression (Fig. 2). The
mRNA in SVOG cells (6-fold increase after 24 h of reasons for the opposite effects of these cAMP
exposure), but it did not affect VEGF expression by elevating agents in human and bovine species are
these cells (Kisliouk et al., 2003). Another study yet unclear. However, this in vitro system may
employing luteinized human GC had in accordance correspond to luteal cells of CL at an early stage,
demonstrated that hCG up-regulated EG-VEGF/PK- when VEGF levels are high and EG-VEGF/PK-1
1 mRNA (Fraser et al., 2005). In contrast, incuba- expression still low (Fraser et al., 2005). Since
tion of bovine granulosa cells for 2–6 days in EG-VEGF/PK-1 synthesis increased in the mature
luteinization promoting medium consisting of for- luteal tissue, it was thought that increasing levels
skolin and insulin inhibited EG-VEGF/PK-1 while of progesterone might stimulate the synthesis of
EG-VEGF/PK-1. However, while progesterone ele-
vated EG-VEGF/PK-1 mRNA levels in human en-
0.6 dometrial tissue (Battersby et al., 2004) it did not
in luteinized granulosa cells (Fraser et al., 2005).
Hypoxia is generally considered to be a primary
0.5
trigger of new blood vessels formation in normal
EG-VEGF/PK-1 mRNA levels

-FI and pathologic tissues (Harris, 2002; Choi et al.,


0.4 +FI 2003). As expected VEGF, both its mRNA and
(Arbitrary units)

protein secreted into the culture media were


0.3 elevated in SVOG cells incubated with hypoxia
mimicking agents as CoCl2 and deferoxamine
mesylate (DFX), but expression of EG-VEGF/PK-1
0.2
was inhibited under the same conditions (Kisliouk
* et al., 2003). These findings contrast those re-
0.1 ported by LeCouter et al. using human adrenal
carcinoma cell lines SW13 and H295R, where both
0 VEGF and EG-VEGF/PK-1 were increased by hypoxia
2d 4d 6d (LeCouter et al., 2001). Whether or not the
different cell types may account for this discre-
100
* pancy is unknown at present.
90 * Thrombin is yet another potent activator of
angiogenesis (Tsopanoglou et al., 2004). Moreover
80
* it up-regulates VEGF receptors in ECs and induces
70 VEGF expression in several cell types (Maragoudakis
VEGF mRNA levels
(Arbitrary units)

and Tsopanoglou, 2000; Bassus et al., 2001; Lock-


60
wood et al., 2002; Wang et al., 2002). Thrombin
50 elevated VEGF expression in SVOG cells while EG-
VEGF/PK-1 expression was downregulated (Kisliouk
40
et al., 2003). In accordance increasing serum
30 concentrations up to 10% also inhibited EG-VEGF/
PK-1 expression (Kisliouk et al., 2003).
20

10

0
2d 4d 6d
Ovarian expression of PK-R1 and PK-R2
and their regulation
Figure 2. Inverse regulation of EG-VEGF/PK-1 (A) and
VEGF (B) mRNA expression in bovine granulosa cells. The Acting via two closely related G protein-coupled
cells were incubating for 2–6 days (D) in basal medium receptors, PK-R1 and PK-R2, PKs peptides regulate
(containing 1% fetal bovine serum, FBS) or in a luteiniza-
diverse biological functions. However, at present it
tion promoting medium (consisting of forskolin (F) and
insulin (I) in basal medium). At each time point RNA was is yet unknown whether these receptors have
extracted and mRNA levels were determined by Real- selective or overlapping functions.
Time RT-PCR. Results are Means 7 SEM from 4 indepen- Expression of PK receptors in heterogeneous
dent experiments. * Denotes significant difference systems showed that they bind to and are activated
(Po0.05) as compared with the matching control time by nanomolar concentrations of recombinant PKs
point. (Lin et al., 2002a; Masuda et al., 2002; Soga et al.,
ARTICLE IN PRESS
Role of prokineticin-1 and its corpus luteum 533

2002). Activation of PK receptors overexpressed in A


cells lead to mobilization of calcium, stimulation of 200
phosphoinoside turnover and activation of p44/p42 180
MAPK cascade (Lin et al., 2002a; Masuda et al.,
2002; Soga et al., 2002). These biological effects 160

(normalized to 0.1% FBS)


PK-R1
were also observed in cells naturally expressing PK-

PK-Rs mRNA levels


140
PK-R2
Rs, namely in ACE (Lin et al., 2002b). 120
We had observed a distinct pattern of PK
100
receptors in bovine ovarian cells. Follicular and
luteal steroidogenic cells predominantly ex- 80
press PK-R1, whereas CL-derived EC express 60
high levels of both PK-R1 and PK-R2 (Kisliouk
40
et al., 2003). PK-R1 mRNA was also detected
in human luteinized granulosa cells and in SVOG 20
cell line (Kisliouk et al., 2003). Up to now, among 0
the different examined EC types PK-R2 were 0.1% FBS 2.5% FBS 10% FBS 2.5% FBS
+GF
identified only in fenestrated EC, such as those
found in the adrenal cortex, CL, kidney and liver B 120
(Masuda et al., 2002; Kisliouk et al., 2003; LeCouter
et al., 2003a), implying that PK-R2 may confer the 100

(normalized to 0.1% FBS)


selective effects of PK-1 on microvascular EC PK-Rs mRNA levels
functions. 80
Our recent study had demonstrated that mRNA
expression of PK receptors in luteal EC is modulated 60
by various stress factors such as serum starvation,
TNF-a and hypoxia induced by DFX or CoCl2 40
(Kisliouk et al., 2005). All treatments significantly
elevated PK-R2 mRNA expression and did not
20
markedly affected PK-R1 mRNA levels (Kisliouk et
al., 2005). Unlike with EC, hypoxia induced by
0
CoCl2 or DFX augmented PK-R1 mRNA expression in 0.1% FBS EGF IGF-1
SVOG cells (Kisliouk et al., 2003).
The relatively low mRNA level of PK-R2 in luteal Figure 3. Modulation of PK-Rs expression by serum and
growth factors in luteal EC. (A) Effects of FBS and growth
EC incubated in the medium consisting 10%
factor mix on PK-Rs mRNA expression. Cells were
serum could suggest that different growth factors
incubated in medium containing various concentrations
present in serum, such as IGF-I and EGF may inhibit of FBS (10%, 2.5% and 0.1%) for 48 h. The mix of growth
PK-R2 expression in luteal EC (Fig. 3). As depic- factors (GF; Cambrex bioproducts, SingleQuots EGMTM-2)
ted in Fig. 3, IGF-I and EGF inhibited PK-R2 mRNA consisted of VEGF, FGF-b, EGF and IGF-1 was added to
expression by 60% and 30%, respectively; FGF-b medium containing 2.5% FBS (2.5% FBS+GF). (B) Effects of
and VEGF cultured alone did not change PK-R2 EGF and IGF-1 on PK-Rs mRNA expression. EGF and IGF-1
mRNA levels (data not shown), whereas the were added in to the medium containing 0.1% FBS for
presence of all these factors simultaneously in 48 h. PK-R1 and PK-R2 mRNA expression were determined
the culture medium (containing 2.5% serum) by Real-Time RT-PCR. Data are mean 7 range of two
markedly decreased PK-R2 mRNA. These levels separate experiments.
were similar to those obtained with 10% serum
alone (inhibition almost by 90%, Fig. 3A). These
same growth factors slightly influenced PK-R1 Effects of Prokineticins on proliferation
expression in luteal EC (Fig. 3). Therefore, growth and survival of bovine luteal endothelial
factors present in serum exert an inhibitory cells
effect on PK-R2 mRNA, while stress conditions
such as serum deprivation followed by hypoxia or Both EG-VEGF/PK-1 and VEGF resulted in ex-
exposure to TNF-a increased PK-R2 mRNA ex- tensive angiogenesis and cyst formation when
pression in luteal EC (Fig. 4) (Kisliouk et al., delivered into mouse ovaries (LeCouter et al.,
2005). Collectively, these findings tend to suggest 2001). In adrenal cortex-derived EC, EG-VEGF/PK-1
that PK-R2 may have a role in the survival of provoked a rapid phosphorylation of p44/42 MAP
microvascular EC. kinase and DNA synthesis in these cells (Lin et al.,
ARTICLE IN PRESS
534 T. Kisliouk et al.

induced 2-fold increase only (Kisliouk et al.,


PK-R1 2005). Therefore the more pronounced effect of
EG-VEGF/PK-1 on [3H]-thymidine incorporation
during hypoxia is likely due to higher PK-R2 mRNA
Steroidogenic expression under these conditions.
Cell MAPK is a critical element in cell proliferation as
was shown by numerous studies (Guo et al., 1995;
EG-VEGF/PK-1 Seger and Krebs, 1995; Widmann et al., 1999). It
have been demonstrated that both prokineticins
induced the phosphorylation of p44/42 MAPK in
VEGF luteal EC (Kisliouk et al., 2005) as they promoted in
PK-R1 ACE (Lin et al., 2002b). MAPK is one of the signaling
PK-R2 Wellbeing
pathways that are known to induce expression of
Serum
GFs
the oncoproteins c-fos and c-jun, which belong to a
class of immediate early genes are rapidly acti-
Stress vated, usually in a transient fashion, in response to
Endothelial Hypoxia intracellular signaling cascades (Shaulian and Kar-
Cell in, 2002; Sng et al., 2004). EG-VEGF/PK1 is a potent
Serum withdrawal
TNFα inducer of c-fos and c-jun mRNA expression. It
transiently augmented c-fos mRNA expression in
luteal EC with maximum stimulation attained at 1 h
Proliferation & survival (7-fold) (Kisliouk et al., 2005). EG-VEGF/PK1 also
increased c-jun mRNA expression, however it was
Figure 4. Paracrine/autocrine actions of EG-VEGF/PK-1
less pronounced than with c-fos (2-fold increase at
in luteal endothelial and steroidogenic cell types. EG-
1 h) (Kisliouk et al., 2005).
VEGF/PK-1 is expressed by luteal steroidogenic cell
possessing PK-R1 where it augments VEGF mRNA expres- The immediate downstream effectors of PK-Rs
sion. EG-VEGF/PK-1 also acts on luteal EC expressing high are just beginning to be resolved. Pretreatment of
levels of both PK-R1 and PK-R2. Activation of PK-Rs in luteal EC with pertussis toxin (PTX), which speci-
luteal EC augments their proliferation and survival. PK-R2 fically modifies the heterotrimeric G protein Gia,
expression in luteal EC is inhibited by serum and growth blocking signaling pathways of G protein coupled
factors (GFs). Stress conditions such as serum with- receptor involving Gia, did not inhibit the effect of
drawal, hypoxia and TNF-a elevate PK-R2 expression, EG-VEGF/PK-1 on [3H]-thymidine incorporation
whereas mRNA levels of PK-R1 remain unchanged. (Kisliouk et al., 2005). Therefore, Gia may not be
involved in PK-R activation in luteal EC. These
results are in agreement with those previously
2002b). These studies support the concept that EG- reported by Lin et al. (2002a) and Soga et al.
VEGF/PK-1 acts as selective angiogenic growth (2002), but contradict those published by the group
factor for endocrine-gland derived endothelium. led by Ferarra in adrenal gland EC (Lin et al.,
Being a highly vascular endocrine gland, it was of 2002b). Recently, Chen et al. have reported, that
interest to investigate the effects of PK-1 on CL- co-expression of PK-R2 with chimeric G protein
derived EC function. Luteal EC were isolated and (Gqi5), which shifts receptor/Gi coupling to Ca2+
maintained in culture as described by Spanel- mobilization signaling, dramatically increased Ca2+
Borowski and colleagues (Lehmann et al., 2000). mobilization in response to ligand stimulation,
PKs were found to be potent mitogens for luteal EC: whereas co-expression of PK-R1 with Gqi5 did not
they increased [3H]-thymidine incorporation by significantly affect Ca2+ mobilization (Chen et al.,
luteal EC almost to the same level as VEGF (by 2005). These results may imply that GI can be
factors of 1.6, 1.7 and 2 over control, for PK-1, coupled to PK-R2 only. This study also showed PK
PK-2 and VEGF, respectively) (Kisliouk et al., 2005). stimulated cAMP accumulation in PKR-expressing
Cell counts were in agreement with the results of cells (Chen et al., 2005) suggesting that PKRs may
[3H]-Thymidine incorporation assay: incubation be coupled to multiple G proteins as observed with
with PKs and VEGF increased luteal EC numbers other GPCR (Hermans, 2003). These findings may
1.5-fold and 2-fold, respectively (Kisliouk et al., also explain why PKs are multifunctional peptides.
2005). Addition of EG-VEGF/PK-1 with hypoxia In addition to activating luteal EC proliferation,
mimicking agent DFX simultaneously induced a prokineticins were shown to efficiently inhibit
2.5-fold increase in [3H]-thymidine uptake as apoptosis in these cells induced by serum starvation
compared with the control, whereas DFX alone (Kisliouk et al., 2005). The anti-apoptotic PKs
ARTICLE IN PRESS
Role of prokineticin-1 and its corpus luteum 535

effect was demonstrated by three different References


methods: nuclear morphology (DAPI staining),
detection of DNA strand breaks (TUNEL Assay), Al-zi’abi, M.O., Watson, E.D., Fraser, H.M., 2003.
and activation of caspase-3 (Kisliouk et al., 2005). Angiogenesis and vascular endothelial growth factor
Serum starvation of luteal EC typically resulted in expression in the equine corpus luteum. Reproduction
16.572.8% apoptotic nuclei as was detected by 125, 259–270.
DAPI staining and 19.975.6% apoptotic cells as was Augustin, H.G., Braun, K., Telemenakis, I., Modlich, U.,
assessed by flow cytometry detection of TUNEL- Kuhn, W., 1995. Ovarian angiogenesis. Phenotypic
positive cells. EG-VEGF/PK-1 and PK-2 were able to characterization of endothelial cells in a physiological
model of blood vessel growth and regression. Am. J.
rescue the cells, and reduced the apoptosis rate to
Pathol. 147, 339–351.
7.071.5% or 8.071.2%, respectively, as detected
Bassus, S., Herkert, O., Kronemann, N., Gorlach, A.,
by DAPI staining and to 4.372.3% and 4.371.9%, Bremerich, D., Kirchmaier, C.M., Busse, R., Schini-
respectively, as measured by TUNEL assay (Kisliouk Kerth, V.B., 2001. Thrombin causes vascular endothe-
et al., 2005). The amount of activated caspase-3 lial growth factor expression in vascular smooth
was determined by western blot analysis using muscle cells: role of reactive oxygen species. Arter-
specific antibody to cleaved caspase-3 (17 and ioscler. Thromb. Vasc. Biol. 21, 1550–1555.
19 kDa). Both PKs, like VEGF, completely inhibited Battersby, S., Critchley, H.O., Morgan, K., Millar, R.P.,
activation of caspase-3 induced by serum starvation Jabbour, H.N., 2004. Expression and regulation of the
(Kisliouk et al., 2005). Consequently, PKs are not prokineticins (endocrine gland-derived vascular en-
only mitogens, but are also survival factors for dothelial growth factor and Bv8) and their receptors in
luteal EC. the human endometrium across the menstrual cycle.
J. Clin. Endocrinol. Metab. 89, 2463–2469.
Besides the mitogenic and anti-apoptotic effects
Berisha, B., Schams, D., Kosmann, M., Amselgruber, W.,
of prokineticins on luteal EC, EG-VEGF/PK-1 ele-
Einspanier, R., 2000. Expression and tissue concentra-
vated VEGF mRNA expression in bovine luteal
tion of vascular endothelial growth factor, its recep-
steroidogenic cells (Kisliouk et al., 2005). Augmen- tors, and localization in the bovine corpus luteum
tation of VEGF expression in luteal steroidogenic during estrous cycle and pregnancy. Biol. Reprod. 63,
cells imply that EG-VEGF/PK-1 could also indirectly, 1106–1114.
via VEGF, affect luteal angiogenesis. Carambula, S.F., Pru, J.K., Lynch, M.P., Matikainen, T.,
Goncalves, P.B., Flavell, R.A., Tilly, J.L., Rueda, B.R.,
2003. Prostaglandin F2alpha- and FAS-activating anti-
body-induced regression of the corpus luteum involves
Conclusions caspase-8 and is defective in caspase-3 deficient mice.
Reprod. Biol. Endocrinol. 1, 15.
EG-VEGF/PK-1 is expressed in human and bovine Chen, J., Kuei, C., Sutton, S., Wilson, S., Yu, J., Kamme,
ovaries where it elicits cell-specific effects in luteal F., Mazur, C., Lovenberg, T., Liu, C., 2005. Identifica-
tion and Pharmacological Characterization of Prokine-
steroidogenic cells and in endothelial cells suggest-
ticin 2{beta} as a Selective Ligand for Prokineticin
ing that EG-VEGF/PK1 may be a novel autocrine/ Receptor 1. Mol. Pharmacol. 67, 2070–2076.
paracrine regulator within the ovary (Fig. 4). PKs Cheng, M.Y., Bullock, C.M., Li, C., Lee, A.G., Bermak,
promote DNA synthesis in adrenal cortex EC, as J.C., Belluzzi, J., Weaver, D.R., Leslie, F.M., Zhou,
they do in luteal EC. These cells have much in Q.Y., 2002. Prokineticin 2 transmits the behavioural
common: irrigating endocrine glands the fene- circadian rhythm of the suprachiasmatic nucleus.
strated microvascular EC are particularly perme- Nature 417, 405–410.
able to the inflow of blood-borne substances and Choi, K.S., Bae, M.K., Jeong, J.W., Moon, H.E., Kim,
the outflow of specific secreted products. It is K.W., 2003. Hypoxia-induced angiogenesis during
noteworthy that adrenal cortex and luteal EC, carcinogenesis. J. Biochem. Mol. Biol. 36, 120–127.
unlike the EC derived from the bovine aorta, Ferrara, N., Frantz, G., LeCouter, J., Dillard-Telm, L.,
equally express the two PK receptor types -R1 and Pham, T., Draksharapu, A., Giordano, T., Peale, F.,
R2 (Fig. 4). PKs also affect endocrine cells: PK-R1 2003. Differential expression of the angiogenic factor
genes vascular endothelial growth factor (VEGF)
were identified in ovarian steroidogenic cells where
and endocrine gland-derived VEGF in normal and
EG-VEGF/PK-1 enhances VEGF mRNA expression.
polycystic human ovaries. Am. J. Pathol. 162,
The presence of PK-R1 and inverse regulation of 1881–1893.
EG-VEGF/PK1 and VEGF mRNA expression by Fraser, H.M., Lunn, S.F., 2001. Regulation and manipula-
ovarian steroidogenic cells suggest that in addition tion of angiogenesis in the primate corpus luteum.
to its angiogenic effects, EG-VEGF/PK-1 may also Reproduction 121, 355–362.
have other functions in ovary, which remain to be Fraser, H.M., Wulff, C., 2003. Angiogenesis in the corpus
investigated. luteum. Reprod. Biol. Endocrinol. 1, 88.
ARTICLE IN PRESS
536 T. Kisliouk et al.

Fraser, H.M., Bell, J., Wilson, H., Taylor, P.D., Morgan, K., Li, M., Bullock, C.M., Knauer, D.J., Ehlert, F.J., Zhoum,
Anderson, R.A., Duncan, W.C., 2005. Localization and Q.Y., 2001. Identification of two prokineticin cDNAs:
quantification of cyclic changes in the expression of recombinant proteins potently contract gastro-
endocrine gland vascular endothelial growth factor in intestinal smooth muscle. Mol. Pharmacol. 59,
the human corpus luteum. J. Clin. Endocrinol. Metab. 692–698.
90, 427–434. Lin, D.C., Bullock, C.M., Ehlert, F.J., Chen, J.L., Tian, H.,
Guo, D., Jia, Q., Song, H.Y., Warren, R.S., Donner, D.B., Zhou, Q.Y., 2002a. Identification and molecular
1995. Vascular endothelial cell growth factor pro- characterization of two closely related G protein-
motes tyrosine phosphorylation of mediators of signal coupled receptors activated by prokineticins/endo-
transduction that contain SH2 domains. Association crine gland vascular endothelial growth factor. J. Biol.
with endothelial cell proliferation. J. Biol. Chem. 270, Chem. 277, 19276–19280.
6729–6733. Lin, R., LeCouter, J., Kowalski, J., Ferrara, N., 2002b.
Harris, A.L., 2002. Hypoxia—a key regulatory factor in Characterization of endocrine gland-derived vascular
tumour growth. Nat. Rev. Cancer 2, 38–47. endothelial growth factor signaling in adrenal cortex
Hazzard, T.M., Molskness, T.A., Chaffin, C.L., Stouffer, capillary endothelial cells. J. Biol. Chem. 277,
R.L., 1999. Vascular endothelial growth factor (VEGF) 8724–8729.
and angiopoietin regulation by gonadotrophin and Lockwood, C.J., Krikun, G., Koo, A.B., Kadner, S.,
steroids in macaque granulosa cells during the Schatz, F., 2002. Differential effects of thrombin and
peri-ovulatory interval. Mol. Hum. Reprod. 5, hypoxia on endometrial stromal and glandular epithe-
1115–1121. lial cell vascular endothelial growth factor expression.
Hermans, E., 2003. Biochemical and pharmacological J. Clin. Endocrinol. Metab. 87, 4280–4286.
control of the multiplicity of coupling at G-protein- Maragoudakis, M.E., Tsopanoglou, N.E., 2000. On the
coupled receptors. Pharmacol. Ther. 99, 25–44. mechanism(s) of thrombin induced angiogenesis. Adv.
Kaser, A., Winklmayr, M., Lepperdinger, G., Kreil, G., Exp. Med. Biol. 476, 47–55.
2003. The AVIT protein family. Secreted cysteine-rich Martinez-Chequer, J.C., Stouffer, R.L., Hazzard, T.M.,
vertebrate proteins with diverse functions. EMBO Patton, P.E., Molskness, T.A., 2003. Insulin-like growth
Reprod. 4, 469–473. factors-1 and -2, but not hypoxia, synergize with
Kisliouk, T., Levy, N., Hurwitz, A., Meidan, R., 2003. gonadotropin hormone to promote vascular endothe-
Presence and regulation of endocrine gland vascular lial growth factor-A secretion by monkey granulosa
endothelial growth factor/prokineticin-1 and its re- cells from preovulatory follicles. Biol. Reprod. 68,
ceptors in ovarian cells. J. Clin. Endocrinol. Metab. 1112–1118.
88, 3700–3707. Masuda, Y., Takatsu, Y., Terao, Y., Kumano, S., Ishibashi,
Kisliouk, T., Podlovni, H., Spanel-Borowski, K., Ovadia, Y., Suenaga, M., Abe, M., Fukusumi, S., Watanabe, T.,
O., Zhou, Q.Y., Meidan, R., 2005. Prokineticins Shintani, Y., Yamada, T., Hinuma, S., Inatomi, N.,
(endocrine gland-VEGF and BV8) in the bovine ovary: Ohtaki, T., Onda, H., Fujino, M., 2002. Isolation and
expression and role as mitogens and survival factors identification of EG-VEGF/prokineticins as cognate
for corpus luteum derived-endothelial cells. Endocri- ligands for two orphan G-protein-coupled receptors.
nology, June 2 (e-pub ahead of press). Biochem. Biophys. Res. Commun. 293, 396–402.
LeCouter, J., Kowalski, J., Foster, J., Hass, P., Zhang, Z., McCracken, J.A., Custer, E.E., Lamsa, J.C., 1999.
Dillard-Telm, L., Frantz, G., Rangell, L., DeGuzman, Luteolysis: a neuroendocrine-mediated event. Physiol.
L., Keller, G.A., Peale, F., Gurney, A., Hillan, K.J., Rev. 79, 263–323.
Ferrara, N., 2001. Identification of an angiogenic Niswender, G.D., Juengel, J.L., Silva, P.J., Rollyson,
mitogen selective for endocrine gland endothelium. M.K., McIntush, E.W., 2000. Mechanisms controlling
Nature 412, 877–884. the function and life span of the corpus luteum.
LeCouter, J., Lin, R., Frantz, G., Zhang, Z., Hillan, K., Physiol. Rev. 80, 1–29.
Ferrara, N., 2003a. Mouse endocrine gland-derived Redmer, D.A., Doraiswamy, V., Bortnem, B.J., Fisher, K.,
vascular endothelial growth factor: a distinct expres- Jablonka-Shariff, A., Grazul-Bilska, A.T., Reynolds,
sion pattern from its human ortholog suggests differ- L.P., 2001. Evidence for a role of capillary pericytes in
ent roles as a regulator of organ-specific angiogenesis. vascular growth of the developing ovine corpus
Endocrinology 144, 2606–2616. luteum. Biol. Reprod. 65, 879–889.
LeCouter, J., Lin, R., Tejada, M., Frantz, G., Peale, F., Schams, D., Berisha, B., 2002. Steroids as local regulators
Hillan, K.J., Ferrara, N., 2003b. The endocrine-gland- of ovarian activity in domestic animals. Domest. Anim.
derived VEGF homologue Bv8 promotes angiogenesis in Endocrinol. 23, 53–65.
the testis: localization of Bv8 receptors to endothelial Schams, D., Berisha, B., 2004. Regulation of corpus
cells. Proc. Natl. Acad. Sci. USA. 100, 2685–2690. luteum function in cattle-an overview. Reprod. Do-
Lehmann, I., Brylla, E., Sittig, D., Spanel-Borowski, K., mest. Anim. 39, 241–251.
Aust, G., 2000. Microvascular endothelial cells differ Schams, D., Berisha, B., Kosmann, M., Amselgruber,
in their basal and tumour necrosis factor-alpha- W.M., 2002. Expression and localization of IGF family
regulated expression of adhesion molecules and members in bovine antral follicles during final growth
cytokines. J. Vasc. Res. 37, 408–416. and in luteal tissue during different stages of estrous
ARTICLE IN PRESS
Role of prokineticin-1 and its corpus luteum 537

cycle and pregnancy. Domest. Anim. Endocrinol. 22, Wang, Z., Tamura, K., Yoshie, M., Tamura, H., Imakawa,
51–72. K., Kogo, H., 2003. Prostaglandin F2alpha-induced
Seger, R., Krebs, E.G., 1995. The MAPK signaling cascade. functional regression of the corpus luteum and
Faseb J. 9, 726–735. apoptosis in rodents. J. Pharmacol. Sci. 92, 19–27.
Shaulian, E., Karin, M., 2002. AP-1 as a regulator of cell Webb, R., Woad, K.J., Armstrong, D.G., 2002. Corpus
life and death. Nat. Cell. Biol. 4, E131–E136. luteum (CL) function: local control mechanisms.
Smith, M.F., McIntush, E.W., Smith, G.W., 1994. Mechan- Domest. Anim. Endocrinol. 23, 277–285.
isms associated with corpus luteum development. J. Wechselberger, C., Puglisi, R., Engel, E., Lepperdinger,
Anim. Sci. 72, 1857–1872. G., Boitani, C., Kreil, G., 1999. The mammalian
Sng, J.C., Taniura, H., Yoneda, Y., 2004. A tale of early homologues of frog Bv8 are mainly expressed in
response genes. Biol. Pharm. Bull. 27, 606–612. spermatocytes. FEBS Lett. 462, 177–181.
Soga, T., Matsumoto, S., Oda, T., Saito, T., Hiyama, H., Widmann, C., Gibson, S., Jarpe, M.B., Johnson, G.L.,
Takasaki, J., Kamohara, M., Ohishi, T., Matsushime, 1999. Mitogen-activated protein kinase: conservation
H., Furuichi, K., 2002. Molecular cloning and char- of a three-kinase module from yeast to human.
acterization of prokineticin receptors. Biochim. Bio- Physiol. Rev. 79, 143–180.
phys. Acta. 1579, 173–179. Wulff, C., Wilson, H., Largue, P., Duncan, W.C.,
Sugino, N., Kashida, S., Takiguchi, S., Karube-Harada, A., Armstrong, D.G., Fraser, H.M., 2000. Angiogenesis in
Kato, H., 2001. Expression of vascular endothelial the human corpus luteum: localization and changes in
growth factor (VEGF) receptors in rat corpus luteum: angiopoietins, tie-2, and vascular endothelial growth
regulation by oestradiol during mid-pregnancy. Repro- factor messenger ribonucleic acid. J. Clin. Endocrinol.
duction 122, 875–881. Metab. 85, 4302–4309.
Tanaka, J., Acosta, T.J., Berisha, B., Tetsuka, M., Matsui, Wulff, C., Wilson, H., Rudge, J.S., Wiegand, S.J., Lunn,
M., Kobayashi, S., Schams, D., Miyamoto, A., 2004. S.F., Fraser, H.M., 2001. Luteal angiogenesis: preven-
Relative changes in mRNA expression of angiopoietins tion and intervention by treatment with vascular
and receptors tie in bovine corpus luteum during endothelial growth factor trap(A40). J. Clin. Endocri-
estrous cycle and prostaglandin F2alpha-induced nol. Metab. 86, 3377–3386.
luteolysis: a possible mechanism for the initiation of Young, F.M., Rodger, F.E., Illingworth, P.J., Fraser, H.M.,
luteal regression. J. Reprod. Dev. 50, 619–626. 2000. Cell proliferation and vascular morphology in
Tsopanoglou, N.E., Papaconstantinou, M.E., Flordellis, the marmoset corpus luteum. Hum. Reprod. 15,
C.S., Maragoudakis, M.E., 2004. On the mode of 557–566.
action of thrombin-induced angiogenesis: thrombin Zetser, A., Kisliouk, T., Ivakin, E., Lahav, M., 2001.
peptide, TP508, mediates effects in endothelial cells Dependence on prolactin of the luteolytic effect of
via alphavbeta3 integrin. Thromb. Haemost. 92, prostaglandin F2alpha in rat luteal cell cultures. Biol.
846–857. Reprod. 65, 1082–1091.
Wang, J., Morita, I., Onodera, M., Murota, S.I., 2002. Zheng, J., Redmer, D.A., Reynolds, L.P., 1993. Vascular
Induction of KDR expression in bovine arterial en- development and heparin-binding growth factors in
dothelial cells by thrombin: involvement of nitric the bovine corpus luteum at several stages of the
oxide. J. Cell. Physiol. 190, 238–250. estrous cycle. Biol. Reprod. 49, 1177–1189.

You might also like