You are on page 1of 8

REVIEW

Cardiovascular Research (2011) 92, 191198


doi:10.1093/cvr/cvr228

Prokineticin receptor 1 (PKR1) signalling


in cardiovascular and kidney functions
Mounia Boulberdaa, Kyoji Urayama, and Canan G. Nebigil*
CNRS, Universite de Strasbourg, UMR7242, Ecole Superieure de Biotechnologie de Strasbourg, ESBS, Boulevard Sebastien Brandt BP 10413, F-67412 Illkirch, France
Received 30 May 2011; revised 9 August 2011; accepted 16 August 2011; online publish-ahead-of-print 19 August 2011

Abstract

----------------------------------------------------------------------------------------------------------------------------------------------------------Keywords

Receptor Prokineticin Cardiomyopathy Renal physiology Progenitor cell

1. Introduction
Prokineticins (PK1 and PK2) are structural homologues of amphibian
or reptilian peptide toxins that were first identified in the gastrointestinal tract1 as potent agents mediating muscle contraction, and have
been isolated from bovine milk.2 They comprise two classes: prokineticin 1 (PK1), originally called endocrine gland-derived vascular endothelial growth factor (EG-VEGF)1 and prokineticin 2 (PK2, also called
Bv8). PK1 and PK2 are approximately 50% homologous to each other
and contain carboxyl terminal cysteine-rich domains that form five
disulfide bridges.3 The N-terminal hexapeptide (AVITGA) and
cysteine residues in the carboxyl terminal domain are crucial for
their biological activities. PK1 is encoded by a gene mapped on
chromosome 1 (NCBI Gene ID: 84432). PK2 is localized on
chromosome 3p13 and contains four exons (NCBI Gene ID:
60675). The most active form of PK2, with 81 amino acids, is
encoded by the exons 1, 2 and 4;3 however, a long form of the
PK2 peptide, PK2L, with 21 additional amino acids, is encoded by all
four exons of PK2. Prokineticins and their receptors are widely distributed in mammalian tissues.4 Regulators for the prokineticins
have been described in reproductive tract, neurons and macrophages.

In reproductive tract, PK1 expression was found to be up-regulated by


estrogen, progesterone, and human chorionic gonadotrophin, as well
as hypoxia-inducible factor 1a.5,6 In olfactory bulbs, PK2 expression
was elevated by two proneural basic helix loop helix factors (neurogenin1 and Mash1) and repressed by homeobox transcriptional
factors (distal-less homeobox 1 and 2).7,8 Transcription factor activating protein-1 is involved in regulation of the expression of the Bv8
homologue in amphibians.9 PK2 was shown to be positively regulated
in CD11b+ Gr1+ myeloid cells, specifically by granulocyte colony
stimulating factor.10 Although this information does not explain the
prevalence of distribution of prokineticins in tissues and the stimuli
that regulate their expression, it is likely that circadian stimuli11 in
the central nervous system and remodelling stimuli12 in the heart
regulate the expression of PK2.
Prokineticins have been shown to modulate cell survival,13 17 cell
motility,18 20 cell excitability,21 24 and other more complex behaviours,25 27 as summarized in Table 1.
Prokineticins bind to two cognate 7-transmembrane G-proteincoupled receptors. Prokineticin receptor 1 (PKR1) and PKR2 share
about 85% amino acid identity and are encoded within distinct
chromosomes in both mice and humans.1 PKR1 is located on

* Corresponding author. Tel: +33 368 854 756, Fax: +33 368 854 829, Email: nebigil@unistra.fr
Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: journals.permissions@oup.com.

Downloaded from by guest on March 26, 2015

Prokineticins (PK1 and PK2) are peptide hormones that exert their biological activity via two common G-proteincoupled receptors: prokineticin receptor (PKR) 1 and 2. Their physiology was originally explored mostly in the
context of angiogenic actions in the reproductive tract and gut motility. Since autocrine and paracrine loops have
been established between PK2 and PKR1 in the heart, in this review we focus on the PK2/PKR1 signalling in the functions of the heart and kidney. PKR1 signalling is required for cardiomyocyte survival and angiogenesis. In the mouse
model of myocardial infarction, intracardiac transient PKR1 transfection protects the structure and function of the
heart. Gain- and loss-of-function studies reveal that PKR1 in mouse heart up-regulates its own ligand and PK2,
which in turn acts as a paracrine signal and promotes epicardin-positive progenitor cell differentiation into a vasculogenic cell type. Transgenic mice over-expressing PKR1 in cardiomyocytes exhibit increased neovascularization. Loss
of PKR1 causes structural and functional changes in the heart and kidney. In isolated epicardin-positive progenitor
cells from the kidney, PK2, acting via PKR1, stimulates differentiation of these progenitor cells into endothelial and
smooth muscle cells. Taken together, these data show that PK2/PKR1 is involved in postnatal cardiac and renal neovascularization. The knowledge gained from these studies should facilitate the discovery of therapeutic interventions
in heart and kidney diseases targeting PKR1.

192

M. Boulberdaa et al.

Table 1 Prokineticins are involved in diverse cellular


activities, including cell survival, motility and excitability,
and complex behaviours
Role

Cell type/function

Cell survival

Endothelial cells13,14
Neuronal cells15
Haematopoietic cells16
Cardiomyocytes12,17

Cell motility

Angiogenesis18
Neurogenesis15
Haematopoiesis19
Neovasculogenesis20

Cell excitability

Gut spasmogen2,21
Pain sensitization22
Circadian rhythm23
Sleep24

Complex behaviours

Feeding25
Drinking26
Anxiety27

....................................................................................

2. The PK2/PKR1 signalling pathway


is an important cardiovascular
regulatory pathway
PK2 appears to be an ideal hormonal candidate to regulate cardiovascular
function. Prokineticins are potent angiogenic factors that have beneficial
effects on cardiac tissue repair, possibly by inducing angiogenesis to
improve coronary circulation. Prokineticins utilize mainly Gq protein signalling for their biological effects. Gaq/Ga11 signalling is an essential
pathway to induce cardiac development32 and hypertrophy.33 Moreover,
Gaq/Ga13 signalling is involved in vessel formation,34 as observed in VEGF
knock-out mice,35 suggesting that prokineticin receptor signalling may act
in concert with other known angiogenic factors in cardiovascular physiology. Prokineticins can act as cytokines,36 inducing differentiation of
murine and human bone marrow cells into the monocyte/macrophage
lineage and activation of monocyte proliferation and differentiation,37

3. Prokineticin receptor 1
expression in cardiovascular
and renal tissues and cells
Both PK2 and PKR1 are expressed in the H9c2 cardioblast cell line
derived from rat heart tissue, as well as in H5V capillary endothelial
cells derived from mouse heart tissue, and in human and mouse
hearts.12,41 In situ hybridization analysis of mouse heart and aorta
samples has shown that PKR1 is expressed in the ventricular wall of
the heart and aortic endothelial cells, but not in smooth muscle
cells of the aorta. Endothelial cells from bovine aorta have also
been shown to express PKR1.42 Immunohistochemical analysis confirmed the presence of PKR1 protein in myocardium and epicardium,
in agreement with the PKR1 transcript expression profile. Together,
these data show that PKR1 and its ligand are expressed postnatally
in cardiovascular tissues and cells and might be functional in the
heart (Figure 1A).12 In the kidney, glomeruli and blood vessels were
strongly labelled with an antibody directed against PKR1. PKR1 is
expressed in endothelial cells within the glomeruli, but not in podocytes and mesangial cells (Figure 1B). PKR1 expression was also
detected in the tubular structures. In situ hybridization confirmed
PKR1 expression in glomerular and tubular structures.43

4. Prokineticin receptor 1 signalling


in cardiovascular cells
Prokineticins serve as mitogens and survival factors for corpus
luteum-derived endothelial cells and bovine aortic endothelial
cells.42 PKR1, acting via Ga11, activates both mitogen-activated
protein kinase and Akt signalling pathways to stimulate proliferation,
migration, and angiogenesis in capillary endothelial cells derived
from heart (H5V cells).28 Activation or over-expression of PKR1 in
isolated cardiomyocytes prevents the apoptosis induced by oxidative
stress by inducing Akt phosphorylation. Gq-coupled receptor activation in isolated cardiomyocytes could promote a survival pathway
by activating Akt signalling.12 Previously, excessive expression of Gq
protein in cardiomyocytes has been shown to induce hypertrophy.44
However, increased Gq protein activity in cardiomyocytes induces
anti-apoptotic signals by transactivating epidermal growth factor
receptor and by Akt activation independent of the ability of Gq signalling to elicit hypertrophy.45 These data show that survival and

Downloaded from by guest on March 26, 2015

chromosome 2p13.1 and PKR2 is located on chromosome 20p12.3.28


In situ analyses indicate that PKR2 is the dominant receptor in the
adult brain, particularly in the hypothalamus, the olfactory ventricular
regions, and the limbic system, whereas PKR1 is widely distributed in
the periphery. Furthermore, in enteric neuronal crest cells, PKR1
expression was up-regulated by activation of the mitogen-activated
protein kinase pathway, probably through c-Jun/Fos transcription
factors,19 where PKR1 works in a co-ordinate fashion with glial
cell line-derived neurotrophic factor to mediate proliferation and
differentiation of enteric neural crest cells.29
Prokineticin receptors activate multiple signalling pathways. The
activation of PKRs stimulates calcium mobilization,4 calcineurin/
nuclear factor of activated t-cells (NFAT) signalling,30 phosphoinositol
turnover, mitogen-activated protein kinase and Akt activation,12 and
zonula occludens-1 (ZO-1), the tight junction associated signalling
proteins, down-regulation.31 PKRs can activate transient receptor
potential vanilloid 1 channels, via activation of protein kinase C,
causing heat-induced hyperalgesia.15

and macrophage migration.38,39 Several cytokines involved in macrophage


proliferation and differentiation, such as macrophage colony-stimulating
factor37 and granulocyte colony-stimulating factor, have beneficial
effects on cardiac remodelling following myocardial infarction.40
Reduced PK2 mRNA and protein levels associated with reduced levels
of its receptor, PKR1, were found in human end-stage failing heart
samples (explanted) compared with non-failing myocardium.12 The differences in PK1 and PKR2 levels between failing and non-failing human hearts
were not significant; however, reductions in PK2 and its receptor, PKR1,
were significant, indicating that angiogenic PKR1 pathology associated
with reduced capillary density in patients with heart failure might lead
to impaired contractility. Moreover, autocrine and paracrine loops have
been shown when PK2 secreted by cardiomyocytes binds and stimulates
PKR1 on cardiomyocyte12 and epicardial progenitor cell surfaces.20 Thus,
PK2/PKR1 signalling may have an important role in heart function.

193

Prokineticin receptor 1 in heart and kidney functions

Figure 1 Expression profile of prokineticin receptor 1 (PKR1). (A) PKR1 is expressed in cardiomyocytes, endothelial cells (ECs), and epicardinpositive progenitor cells (EPPCs), but not in vascular smooth muscle cells (SMCs). (B) In the kidney, PKR1 is expressed in glomerular endothelial
cells (GECs), interstitial capillary endothelial cells (ICEs), and tubules, but not in mesangial cells and podocytes.

hypertrophic responses to Gaq are mediated by different signalling


pathways. Given the known functions of Akt,46 these data are consistent with Akt having a central role in PKR1 signalling for protection of
cardiomyocytes against apoptosis.

Myocardial infarction (MI) triggers inflammation, scar formation, and


scar remodelling, which are crucial for tissue repair.47 Following MI,
chemokines induce migration of the circulating blood monocytes
into the infarcted myocardium, where they can differentiate into
macrophages. Macrophage infiltration into the left ventricle (LV) promotes phagocytosis, wound debridement, fibroblast activation and
proliferation, collagen metabolism, and angiogenesis, which have a
major role in the wound-healing process.48 Macrophages are involved
in removal of necrotic cardiac myocytes and apoptotic neutrophils;
secretion of cytokines, chemokines, and growth factors; and modulation of the angiogenic response.49 Macrophage depletion impairs
wound healing and increases LV remodelling following myocardial
injury in mice.50 Indeed, it has been shown in early clinical trials that
inhibition of the inflammatory component with methylprednisolone
drastically increased mortality due to LV rupture.51
Prokineticins are described as chemokines/cytokines for monocytes
and macrophages.36 Prokineticins and their receptors are highly
expressed in inflamed tissues associated with infiltrating cells.52 In
addition, PK2 is up-regulated at the site of rupture of abdominal aortic
aneurysms,53 and in peripheral monocytes and neutrophils in response
to granulocyte colony-stimulating factor.54 Interestingly, expression of
PK2 and its receptors was significantly increased within 48 h and
remained elevated 1 week after coronary ligation in a mouse MI
model (Nebigil laboratory, C. G. Nebigil and K. Urayama, unpublished
observations). Moreover, a 30% increase of phosphorylated Akt correlated with increased levels of PK2 in ischaemic hearts, initializing the
endogenous wound-healing process via prokineticin signalling.12 Intracardiac PKR1 gene transfer utilizing adenovirus after MI in mice further
increased PKR1 levels by approximately four times within 24 h.12 PKR1
gene transfer resulted in a reduction of the MI size, improvement of
left ventricular performance, and consequently, a reduction of mortality

6. Prokineticin receptor 1
in cardiovascular and renal
pathophysiology
Recently, it has been demonstrated that conditional disruption of the
PKR1 gene in mice provokes heart and kidney disorders, including

Downloaded from by guest on March 26, 2015

5. Prokineticin signalling
in myocardial infarction

compared with untreated control mice. Neither VEGF protein levels nor
VEGF A transcripts 164 and 188 were altered in PKR1-treated hearts
compared with vehicle-treated hearts after MI, consistent with the
observation in cultured endothelial cells following prokineticin 2 treatment. These data clearly show that PKR1 gene therapy induces capillary
network growth without increasing VEGF levels. The cardioprotector,
Akt, was found to be increased by 60% in vivo in the PKR1-treated
hearts and PK2-treated cardiomyocytes.12 These data thus suggest that
transient PKR1 gene therapy has beneficial effects on recovery from
myocardial infarction.
What is the mechanism of the cardioprotective effect of PKR1
signalling in the infarcted heart? Transient gene therapy with PKR1
enhanced angiogenesis and reduced apoptosis after MI.12 Prevention
of apoptosis in cardiomyocytes is a possible mechanism of
PKR1-mediated cardioprotective effects. A second mechanism to preserve myocardial function is to promote collateral vessel formation
in order to overcome insufficient tissue oxygenation. Therefore,
PKR1-mediated angiogenesis may be important to maintain sufficient
numbers of living cardiomyocytes and to allow successful cardioprotection following its early phase anti-apoptotic effect. A third mechanism could be that the inflammation itself might contribute to
PKR1-mediated angiogenesis in infarcted hearts. In fact, PKR1 signalling is involved in inflammation, monocyte activation, and macrophage
differentiation,38,39 and furthermore, PKR1 knock-out mice exhibit
lack of inflammation in response to PK2 stimulation.55 However, the
number of infiltrated cells in the scar area after the myocardial infarction was not significantly different between the PKR1 gene-transfected
and control mice following MI, arguing against an indirect effect of
PKR1 through the inflammatory response. A fourth mechanism for
PKR1 to preserve myocardial function, through the induction of
progenitor cell differentiation, remains to be investigated.

194

7. Prokineticin receptor 1 signalling


in progenitor cells derived from
heart and kidney
Cardiac epicardin positive (epicardial derived) progenitor cells
(EPDCs or EPPCs) are characterized as the endogenous progenitor
cells in the heart.62 Moreover, adult EPDCs represent a bona fide
source of cardiovascular progenitor cells, which have the capacity
to respond to multiple cues. A combination of myocardial VEGF
and fibroblast growth factor signalling promotes EPDC differentiation
into endothelial cells. Platelet-derived growth factor, transforming
growth factor b, or bone morphogenetic protein 2 are involved in
differentiation of EPDCs into smooth muscle cells.63 Only two
factors have been identified as capable of stimulating adult EPDCs
into vasculogenic cell types. Thymosin b4 has been described as a

factor that is capable of activating adult EPDCs, resulting in the differentiation of EPDCs into endothelial cells, smooth muscle cells, and
fibroblasts.64 Prokineticin 2 is also able to stimulate adult EPDCs;20
however, prokineticin/PKR1 signalling reprogrammes adult EPDCs
to differentiate only into endothelial and smooth muscle cells, but
not into fibroblasts. Transgenic mice over-expressing PKR1 in the
heart displayed an increased number of epicardin-positive EPDCs,
with an increase of capillary density and coronary vessels. This
mouse model demonstrated a novel PK2/PKR1 signalling pathway
that is involved in communication between cardiomyocytes and
EPDCs, thereby promoting cardiac neovascularization.20
Interestingly, epicardin-positive progenitor cells in the PKR1-null
mutant epicardium, subepicardium, and glomerulus were significantly
lower than those in wild-type tissues.43 PK2 promoted differentiation
of renal epicardin-positive progenitor cells into platelet endothelial
cell adhesion molecule-1-positive endothelial and a-smooth muscle
actin-positive vascular smooth muscle cells, and this effect was
blocked in the PKR1-null mutant renal epicardin-positive progenitor
cells.43 Taken together, these data demonstrate that PKR1 is involved
in regulating postnatal neovasculogenesis in the heart and the kidney
(Figure 2). It is noteworthy that epicardin-positive progenitor cells
resident in both the heart and the kidney express PKR1. Recently, a
relationship between the epicardium and the glomerulus was identified, originating in a similar progenitor tissue,65 and expressing
similar genes, such as podoplanin, a podocytic marker, the T-box
gene Tbx18, and Wt1. 66 Figure 3 summarizes the potential role of
epicardin-positive progenitor cells derived from pro-epicardium in
glomerular and epicardial angiogenesis.

8. Chronic inactivation of PKR1 in


mice produces a phenotype similar
to adverse effects of anti-angiogenic
therapy
Prokineticins are implicated in development of the polycystic ovary
syndrome,67 and of neuroblastoma,27 colorectal, testicular and prostate cancers (summarized in Table 2). It has recently been demonstrated that prokineticin 2 secreted from myeloid cells strongly
induces tumour angiogenesis. Moreover, anti-prokineticin 2 treatment significantly decreases tumour-infiltrating myeloid cell mobilization, thereby inhibiting tumour growth.10 Thus, the use of
immunoneutralizing antibodies or antagonists of prokineticin 2 and

Figure 2 Epicardin-positive progenitor cells derived from the heart and kidney can differentiate into a-smooth muscle actin-positive smooth muscle
cells (SMCs) and platelet endothelial cell adhesion molecule-1-positive endothelial cells (ECs) upon stimulation of PKR1 by its ligand, PK2.

Downloaded from by guest on March 26, 2015

cardiomegaly, interstitial fibrosis and cardiac dysfunction in stress


conditions, renal tubular dilatation, abnormal glomerular capillaries,
an increase in urinary phosphate excretion, and proteinuria.43
PKR1-null mutant mice displayed morphological abnormalities in
both hearts and kidneys at the neonatal stage, eliminating the
possibility of a cardiorenal syndrome.56 Several angiogenic factors
have been shown to regulate the function of the heart and kidney.
Mice lacking platelet-derived growth factor B display renal and
cardiovascular abnormalities.57,58 VEGF has been shown to be
involved in heart development,38 and in glomerulogenesis and
tubulogenesis in the kidney.59 Fibroblast growth factor is also
involved in heart development and function,60 and in glomerulus
formation.61
PKR1-null mutant mice exhibited swollen mitochondria with few
cristae and increased apoptosis in both heart and kidneys. Impaired
capillary formation in mutant heart and kidneys at early ages
creates a hypoxic environment that up-regulates hypoxia-inducible
factor-1a and pro-angiogenic factors as a compensatory mechanism
to restore capillary perturbation at the adult stage. This compensatory recuperation of capillary formation was only observed in the
extraglomerular area; however, a small number of epicardial and glomerular capillary networks remained unaltered, because of increased
apoptosis and reduced progenitor cell numbers found in both tissues.
In summary, the loss of PKR1 causes renal and cardiac structural and
functional changes because of deficits in survival signalling, mitochondrial, and progenitor cell functions in both the heart and the
kidneys.43

M. Boulberdaa et al.

Prokineticin receptor 1 in heart and kidney functions

195

Table 2 Involvement of prokineticins in human diseases


Domain

Role/expression/mutation in human diseases

Reproduction

Endometrial vascular function76


Mid- to late luteal phase77
Ectopic endometrium in endometriosis6
Host implantation during early pregnancy78
Ectopic pregnancy79
Idiopathic recurrent pregnancy loss80
Development of the olfactory system and reproductive
axis in humans81
Mutation in Kallman sydrome and idiopathic
hypogonadotrophic hypogonadism82

Behaviour

Mood disorders in the Japanese population83


Methamphetamine dependence in the Japanese
population84

Cancer

Prostate malignancy85
Neuroblastoma progression86
Angiogenesis in pancreatic disease87
Human tumours and inflammatory disorders88

Cardiovascular

Abdominal aortic aneurysms53


Human end-stage cardiac failure12

....................................................................................

its receptors may be a useful strategy for cancer treatment.


However, anti-angiogenesis treatments targeting platelet-derived
growth factor, VEGF and its receptors or receptor tyrosine kinase
signalling have been associated with clinical renal and cardiac toxicity.68 70 These conditions include cardiac impairment, proteinuria,
hypertension, haemorrhage, and thromobosis, as well as impairment

of wound healing and tissue repair. Although several hypotheses


have been put forward regarding the aetiology of angiogenesis
inhibition-related cardiovascular and renal adverse effects, many of
the underlying pathophysiological mechanisms remain to be elucidated. Some of the adverse effects of anti-angiogenic drugs could
be due to an off-target effect of the drugs.
Do PKR1-null mutant mice display heart and kidney disorders
reminiscent of anti-angiogenic therapy-mediated adverse effects?
Cardiomyopathy, proteinuria, renal disorders, and impaired angiogenesis following rebound neovascularization were evident in
PKR1-null mutant mice. These mice exhibit progressive fibrosis in
their remodelling hearts and kidneys. On the contrary, anti-VEGF
anti-angiogenic therapy reduces fibrosis and collagen deposits in
the lungs and the liver by an unknown mechanism.71 It is not
evident that this reduction in collagen deposits is due to an inhibition of VEGF signalling, because administration of VEGF had a
clear therapeutic benefit, slowing the development of renal fibrosis.72 Neonatal and 3-week-old PKR1 mutant mice have fewer
than normal numbers of capillaries in their hearts and kidneys,
establishing hypoxic conditions with impaired mitochondrial function, as seen in humans and mice treated with anti-angiogenic
drugs.73 This defect in angiogenesis was compensated by an increase
in hypoxia-inducible factor 1a production, accompanying an
increase in pro-angiogenic factors in the mutant heart and kidneys
at the expense of the epicardial and glomerular zone. The activation
and/or up-regulation of other pro-angiogenic signalling pathways as
a form of evasive resistance to angiogenesis inhibitors has been
shown to circumvent anti-angiogenic treatment, triggering neovascularization in due course.74 Restoration of the angiogenic pathways

Downloaded from by guest on March 26, 2015

Figure 3 The relationship between glomerulus and epicardium has been postulated via pro-epicardial cells. Pro-epicardium gives rise to epicardial
progenitor cells. Epicardium has an essential modulating role in the differentiation of the compact ventricular layer of the myocardium and the development of cardiac vessels. Moreover, the epicardial progenitor cells have been shown to differentiate into the pronephric external glomerulus (PEG),
a structure composed of capillary networks, mesangial cells, and podocytes in vertebrates.

196

M. Boulberdaa et al.

Figure 4 We hypothesize that impaired epicardin-positive progenitor cell homing and/or endothelial dysfunction could be involved in heart and
kidney pathology observed in PKR1-null mutant mice.

ischaemic hearts and kidneys. The race is on to facilitate drug


discovery for targets acting on cardiomyocytes or epicardin-positive
progenitor cells to invoke new coronary vessels, and cardiac and
renal tissues as a significant step towards cardioprotection, and
cardiovascular and renal regeneration.

Acknowledgements
We acknowledge all the members of the Nebigil laboratory for their
fruitful discussion during preparation of this article. We wish to thank
to Dr Emel Songu-Mize for her critical reading and discussions.
Conflict of interest: none declared.

Funding

9. Perspectives
Taken together, these data show that PK2, acting via PKR1 signalling,
has important actions on heart and kidney physiology and pathophysiology. Clearly, the main avenues for future studies will be to investigate cell-specific inactivation of PKR1 in a mouse model to further
and fully elaborate the common mechanism underlying pathology in
PKR1 mutant heart and kidneys. More specifically, whether impaired
homing of epicardin-positive progenitor cells and/or endothelial cell
dysfunction in PKR1-null mice leads to heart and kidney pathology
remains to be further investigated (Figure 4). The role of prokineticins
in the pathophysiology of human heart and kidney diseases remains to
be explored. Table 2 summarizes the involvement of prokineticin in
these diseases.76 90
PKR1 is involved in postnatal cardiac and renal vascularization
by activating organ-specific progenitor cells. The identification of
factors that stimulate endogenous cardiac and renal stem or progenitor cells to induce neovascularization and cardiomyocyte/tubular cell
replacement should lead towards therapeutic intervention in heart
and kidney diseases. In this context, prokineticins hold promise for
use as angiogenic factors and for progenitor cell-based therapy in

This work was supported by grants from the Centre National de la


Recherche Scientifique (CNRS), Fondation de France, and Association
Francaise contre les Myopathies (AFM). M.B. and K.U. are supported by
fellowships from AFM and Region Alsace, respectively.

References
1. Li M, Bullock CM, Knauer DJ, Ehlert FJ, Zhou QY. Identification of two prokineticin
cDNAs: recombinant proteins potently contract gastrointestinal smooth muscle.
Mol Pharmacol 2001;59:692 698.
2. Masuda Y, Takatsu Y, Terao Y, Kumano S, Ishibashi Y, Suenaga M et al. Isolation and
identification of EG-VEGF/prokineticins as cognate ligands for two orphan
G-protein-coupled receptors. Biochem Biophys Res Commun 2002;293:396402.
3. Bullock CM, Li JD, Zhou QY. Structural determinants required for the bioactivities of
prokineticins and identification of prokineticin receptor antagonists. Mol Pharmacol
2004;65:582 588.
4. Soga T, Matsumoto S, Oda T, Saito T, Hiyama H, Takasaki J et al. Molecular cloning
and characterization of prokineticin receptors. Biochim Biophys Acta 2002;1579:
173 179.
5. Ngan ES, Lee KY, Yeung WS, Ngan HY, Ng EH, Ho PC. Endocrine gland-derived vascular endothelial growth factor is expressed in human peri-implantation endometrium, but not in endometrial carcinoma. Endocrinology 2006;147:88 95.
6. Tiberi F, Tropea A, Romani F, Apa R, Marana R, Lanzone A. Prokineticin 1, homeobox
A10, and progesterone receptor messenger ribonucleic acid expression in primary
cultures of endometrial stromal cells isolated from endometrium of healthy
women and from eutopic endometrium of women with endometriosis. Fertil Steril
2010;94:2558 2563.

Downloaded from by guest on March 26, 2015

in the PKR1 mutant heart and kidneys could not reverse the damage,
because of defective survival pathways, such as reduced Akt
activity,12,43 increased caspase-3 activity, and impaired common
progenitor cell function in the neonatal mutant hearts and
kidneys. Interestingly, a compensatory mechanism restored the
extraglomerular capillary network without an alteration in the
number of glomerular capillaries in the mutant kidneys. Interstitial
cells and the tubular epithelium also provide signals such as VEGF
for maintenance of the peritubular capillary network, independent
of the glomerular capillary network.75 Our findings clearly demonstrated that the level of VEGF in the glomerulus was not altered,
but VEGF was indeed increased in mutant interstitial cells and
tubular epithelium, possibly restoring extraglomerular angiogenesis.

Prokineticin receptor 1 in heart and kidney functions

34. Moers A, Nieswandt B, Massberg S, Wettschureck N, Gruner S, Konrad I et al. G13 is


an essential mediator of platelet activation in hemostasis and thrombosis. Nat Med
2003;9:1418 1422.
35. Bellomo D, Headrick JP, Silins GU, Paterson CA, Thomas PS, Gartside M et al. Mice
lacking the vascular endothelial growth factor-B gene (Vegfb) have smaller hearts, dysfunctional coronary vasculature, and impaired recovery from cardiac ischemia. Circ Res
2000;86:E29 E35.
36. Monnier J, Samson M. Cytokine properties of prokineticins. FEBS J 2008;275:
4014 4021.
37. Okazaki T, Ebihara S, Asada M, Yamanda S, Saijo Y, Shiraishi Y et al. Macrophage
colony-stimulating factor improves cardiac function after ischemic injury by inducing
vascular endothelial growth factor production and survival of cardiomyocytes. Am J
Pathol 2007;171:1093 1103.
38. Dorsch M, Qiu Y, Soler D, Frank N, Duong T, Goodearl A et al. PK1/EG-VEGF
induces monocyte differentiation and activation. J Leukoc Biol 2005;78:426 434.
39. Martucci C, Franchi S, Giannini E, Tian H, Melchiorri P, Negri L et al. Bv8, the amphibian homologue of the mammalian prokineticins, induces a proinflammatory phenotype of mouse macrophages. Br J Pharmacol 2006;147:225 234.
40. Harada M, Qin Y, Takano H, Minamino T, Zou Y, Toko H et al. G-CSF prevents
cardiac remodeling after myocardial infarction by activating the Jak-Stat pathway in
cardiomyocytes. Nat Med 2005;11:305 311.
41. Parker R, Liu M, Eyre HJ, Copeland NG, Gilbert DJ, Crawford J et al. Y-receptor-like
genes GPR72 and GPR73: molecular cloning, genomic organisation and assignment to
human chromosome 11q21.1 and 2p14 and mouse chromosome 9 and 6. Biochim
Biophys Acta 2000;1491:369 375.
42. Podlovni H, Ovadia O, Kisliouk T, Klipper E, Zhou QY, Friedman A et al. Differential
expression of prokineticin receptors by endothelial cells derived from different vascular beds: a physiological basis for distinct endothelial function. Cell Physiol Biochem
2006;18:315 326.
43. Boulberdaa M, Turkeri G, Urayama K, Dormishian M, Szatkowski C, Zimmer L et al.
Genetic inactivation of prokineticin receptor-1 leads to heart and kidney disorders.
Arterioscler Thromb Vasc Biol 2011;31:842850.
44. Adams JW, Brown JH. G-proteins in growth and apoptosis: lessons from the heart.
Oncogene 2001;20:1626 1634.
45. Howes AL, Miyamoto S, Adams JW, Woodcock EA, Brown JH. Gaq expression activates EGFR and induces Akt mediated cardiomyocyte survival: dissociation from Gaq
mediated hypertrophy. J Mol Cell Cardiol 2006;40:597 604.
46. Nagoshi T, Matsui T, Aoyama T, Leri A, Anversa P, Li L et al. PI3K rescues the detrimental effects of chronic Akt activation in the heart during ischemia/reperfusion
injury. J Clin Invest 2005;115:2128 2138.
47. Jiang B, Liao R. The paradoxical role of inflammation in cardiac repair and regeneration. J Cardiovasc Transl Res 2010;3:410 416.
48. Ren G, Dewald O, Frangogiannis NG. Inflammatory mechanisms in myocardial infarction. Curr Drug Targets Inflamm Allergy 2003;2:242256.
49. van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH, van Luyn MJ. Macrophage depletion impairs wound healing and increases left ventricular remodeling after
myocardial injury in mice. Am J Pathol 2007;170:818829.
50. Frangogiannis NG, Smith CW, Entman ML. The inflammatory response in myocardial
infarction. Cardiovasc Res 2002;53:31 47.
51. Mannisi JA, Weisman HF, Bush DE, Dudeck P, Healy B. Steroid administration after
myocardial infarction promotes early infarct expansion. A study in the rat. J Clin
Invest 1987;79:1431 1439.
52. Catalano RD, Lannagan TR, Gorowiec M, Denison FC, Norman JE, Jabbour HN. Prokineticins: novel mediators of inflammatory and contractile pathways at parturition?
Mol Hum Reprod 2010;16:311 319.
53. Choke E, Cockerill GW, Laing K, Dawson J, Wilson WR, Loftus IM et al. Whole
genome-expression profiling reveals a role for immune and inflammatory response
in abdominal aortic aneurysm rupture. Eur J Vasc Endovasc Surg 2009;37:305310.
54. Kowanetz M, Wu X, Lee J, Tan M, Hagenbeek T, Qu X et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+
granulocytes. Proc Natl Acad Sci USA 2010;107:21248 21255.
55. Giannini E, Lattanzi R, Nicotra A, Campese AF, Grazioli P, Screpanti I et al. The chemokine Bv8/prokineticin 2 is up-regulated in inflammatory granulocytes and modulates inflammatory pain. Proc Natl Acad Sci USA 2009;106:14646 14651.
56. Shamseddin MK, Parfrey PS. Mechanisms of the cardiorenal syndromes. Nat Rev
Nephrol 2009;5:641649.
57. Soriano P. Abnormal kidney development and hematological disorders in PDGF
b-receptor mutant mice. Genes Dev 1994;8:1888 1896.
58. Van den Akker NM, Winkel LC, Nisancioglu MH, Maas S, Wisse LJ, Armulik A et al.
PDGF-B signaling is important for murine cardiac development: its role in developing
atrioventricular valves, coronaries, and cardiac innervation. Dev Dyn 2008;237:
494 503.
59. Villegas G, Lange-Sperandio B, Tufro A. Autocrine and paracrine functions of vascular
endothelial growth factor (VEGF) in renal tubular epithelial cells. Kidney Int 2005;67:
449 457.
60. Marguerie A, Bajolle F, Zaffran S, Brown NA, Dickson C, Buckingham ME et al. Congenital heart defects in Fgfr2-IIIb and Fgf10 mutant mice. Cardiovasc Res 2006;71:
50 60.

Downloaded from by guest on March 26, 2015

7. Long JE, Garel S, Alvarez-Dolado M, Yoshikawa K, Osumi N, Alvarez-Buylla A et al.


Dlx-dependent and -independent regulation of olfactory bulb interneuron differentiation. J Neurosci 2007;27:3230 3243.
8. Zhang C, Ng KL, Li JD, He F, Anderson DJ, Sun YE et al. Prokineticin 2 is a target gene
of proneural basic helix-loop-helix factors for olfactory bulb neurogenesis. J Biol Chem
2007;282:6917 6921.
9. Marsango S, di Patti MC, Barra D, Miele R. The Bv8 gene from Bombina orientalis: molecular cloning, genomic organization and functional characterization of the promoter.
Peptides 2009;30:2182 2190.
10. Shojaei F, Wu X, Zhong C, Yu L, Liang XH, Yao J et al. Bv8 regulates
myeloid-cell-dependent tumour angiogenesis. Nature 2007;450:825 831.
11. Watanabe T, Naito E, Nakao N, Tei H, Yoshimura T, Ebihara S. Bimodal clock gene
expression in mouse suprachiasmatic nucleus and peripheral tissues under a 7-hour
light and 5-hour dark schedule. J Biol Rhythms 2007;22:58 68.
12. Urayama K, Guilini C, Messaddeq N, Hu K, Steenman M, Kurose H et al. The prokineticin receptor-1 (GPR73) promotes cardiomyocyte survival and angiogenesis.
FASEB J 2007;21:2980 2993.
13. Ingves MV, Ferguson AV. Prokineticin 2 modulates the excitability of area postrema
neurons in vitro in the rat. Am J Physiol Regul Integr Comp Physiol 2008;298:R617 R626.
14. Wade PR, Palmer JM, Mabus J, Saunders PR, Prouty S, Chevalier K et al. Prokineticin-1
evokes secretory and contractile activity in rat small intestine. Neurogastroenterol Motil
2009;22:e152 e161.
15. Negri L, Lattanzi R, Giannini E, Colucci M, Margheriti F, Melchiorri P et al. Impaired
nociception and inflammatory pain sensation in mice lacking the prokineticin receptor
PKR1: focus on interaction between PKR1 and the capsaicin receptor TRPV1 in pain
behavior. J Neurosci 2006;26:6716 6727.
16. Li JD, Hu WP, Boehmer L, Cheng MY, Lee AG, Jilek A et al. Attenuated circadian
rhythms in mice lacking the prokineticin 2 gene. J Neurosci 2006;26:11615 11623.
17. Hu WP, Li JD, Zhang C, Boehmer L, Siegel JM, Zhou QY. Altered circadian and
homeostatic sleep regulation in prokineticin 2-deficient mice. Sleep 2007;30:247 256.
18. LeCouter J, Ferrara N. EG-VEGF and the concept of tissue-specific angiogenic growth
factors. Semin Cell Dev Biol 2002;13:3 8.
19. Ngan ES, Lee KY, Sit FY, Poon HC, Chan JK, Sham MH et al. Prokineticin-1 modulates
proliferation and differentiation of enteric neural crest cells. Biochim Biophys Acta 2007;
1773:536 545.
20. LeCouter J, Zlot C, Tejada M, Peale F, Ferrara N. Bv8 and endocrine gland-derived
vascular endothelial growth factor stimulate hematopoiesis and hematopoietic cell
mobilization. Proc Natl Acad Sci USA 2004;101:16813 16818.
21. Urayama K, Guilini C, Turkeri G, Takir S, Kurose H, Messaddeq N et al. Prokineticin
receptor-1 induces neovascularization and epicardial-derived progenitor cell differentiation. Arterioscler Thromb Vasc Biol 2008;28:841849.
22. Gardiner JV, Bataveljic A, Patel NA, Bewick GA, Roy D, Campbell D et al. Prokineticin
2 is a hypothalamic neuropeptide that potently inhibits food intake. Diabetes 2010;59:
397 406.
23. Negri L, Lattanzi R, Giannini E, De Felice M, Colucci A, Melchiorri P. Bv8, the amphibian homologue of the mammalian prokineticins, modulates ingestive behaviour in
rats. Br J Pharmacol 2004;142:181 191.
24. Li JD, Hu WP, Zhou QY. Disruption of the circadian output molecule prokineticin 2
results in anxiolytic and antidepressant-like effects in mice. Neuropsychopharmacology
2009;34:367 373.
25. Kisliouk T, Podlovni H, Spanel-Borowski K, Ovadia O, Zhou QY, Meidan R. Prokineticins (endocrine gland-derived vascular endothelial growth factor and BV8) in the
bovine ovary: expression and role as mitogens and survival factors for corpus luteumderived endothelial cells. Endocrinology 2005;146:3950 3958.
26. Guilini C, Urayama K, Turkeri G, Dedeoglu DB, Kurose H, Messaddeq N et al. Divergent roles of prokineticin receptors in the endothelial cells: angiogenesis and fenestration. Am J Physiol Heart Circ Physiol 2010;298:H844 H852.
27. Nebigil CG. Prokineticin receptors in cardiovascular function: foe or friend? Trends
Cardiovasc Med 2009;19:5560.
28. Kaser A, Winklmayr M, Lepperdinger G, Kreil G. The AVIT protein family. Secreted
cysteine-rich vertebrate proteins with diverse functions. EMBO Rep 2003;4:469 473.
29. Ngan ES, Shum CK, Poon HC, Sham MH, Garcia-Barcelo MM, Lui VC et al.
Prokineticin-1 (Prok-1) works coordinately with glial cell line-derived neurotrophic
factor (GDNF) to mediate proliferation and differentiation of enteric neural crest
cells. Biochim Biophys Acta 2008;1783:467478.
30. Maldonado-Perez D, Brown P, Morgan K, Millar RP, Thompson EA, Jabbour HN. Prokineticin 1 modulates IL-8 expression via the calcineurin/NFAT signaling pathway.
Biochim Biophys Acta 2009;1793:1315 1324.
31. Urayama K, Dedeoglu DB, Guilini C, Frantz S, Ertl G, Messaddeq N et al. Transgenic
myocardial overexpression of prokineticin receptor-2 (GPR73b) induces hypertrophy
and capillary vessel leakage. Cardiovasc Res 2009;81:28 37.
32. Offermanns S, Zhao LP, Gohla A, Sarosi I, Simon MI, Wilkie TM. Embryonic cardiomyocyte hypoplasia and craniofacial defects in Gaq/Ga11-mutant mice. EMBO J 1998;
17:4304 4312.
33. Wettschureck N, Rutten H, Zywietz A, Gehring D, Wilkie TM, Chen J et al. Absence
of pressure overload induced myocardial hypertrophy after conditional inactivation of
Gaq/Ga11 in cardiomyocytes. Nat Med 2001;7:1236 1240.

197

198

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.
89.

90.

factor and Bv8) and their receptors in the human endometrium across the menstrual
cycle. J Clin Endocrinol Metab 2004;89:2463 2469.
Fraser HM, Bell J, Wilson H, Taylor PD, Morgan K, Anderson RA et al. Localization
and quantification of cyclic changes in the expression of endocrine gland vascular
endothelial growth factor in the human corpus luteum. J Clin Endocrinol Metab
2005;90:427 434.
Evans J, Catalano RD, Morgan K, Critchley HO, Millar RP, Jabbour HN. Prokineticin 1
signaling and gene regulation in early human pregnancy. Endocrinology 2008;149:
2877 2887.
Shaw JL, Denison FC, Evans J, Durno K, Williams AR, Entrican G et al. Evidence of
prokineticin dysregulation in fallopian tube from women with ectopic pregnancy.
Fertil Steril 2010;94:1601-8.e1.
Su MT, Lin SH, Lee IW, Chen YC, Hsu CC, Pan HA et al. Polymorphisms of endocrine
gland-derived vascular endothelial growth factor gene and its receptor genes are
associated with recurrent pregnancy loss. Hum Reprod 2010;25:2923 2930.
Dode C, Teixeira L, Levilliers J, Fouveaut C, Bouchard P, Kottler ML et al. Kallmann
syndrome: mutations in the genes encoding prokineticin-2 and prokineticin
receptor-2. PLoS Genet 2006;2:e175.
Cole LW, Sidis Y, Zhang C, Quinton R, Plummer L, Pignatelli D et al. Mutations in
prokineticin 2 and prokineticin receptor 2 genes in human gonadotrophin-releasing
hormone deficiency: molecular genetics and clinical spectrum. J Clin Endocrinol
Metab 2008;93:3551 3559.
Kishi T, Kitajima T, Tsunoka T, Okumura T, Ikeda M, Okochi T et al. Possible association of prokineticin 2 receptor gene (PROKR2) with mood disorders in the Japanese
population. Neuromolecular Med 2009;11:114 122.
Kishi T, Kitajima T, Tsunoka T, Okumura T, Okochi T, Kawashima K et al. PROKR2 is
associated with methamphetamine dependence in the Japanese population. Prog
Neuropsychopharmacol Biol Psychiatry 2010;34:1033 1036.
Pasquali D, Rossi V, Staibano S, De Rosa G, Chieffi P, Prezioso D et al. The
endocrine-gland-derived vascular endothelial growth factor (EG-VEGF)/prokineticin
1 and 2 and receptor expression in human prostate: up-regulation of EG-VEGF/prokineticin 1 with malignancy. Endocrinology 2006;147:4245 4251.
Ngan ES, Sit FY, Lee K, Miao X, Yuan Z, Wang W et al. Implications of endocrine
gland-derived vascular endothelial growth factor/prokineticin-1 signaling in human
neuroblastoma progression. Clin Cancer Res 2007;13:868 875.
Jiang X, Abiatari I, Kong B, Erkan M, De Oliveira T, Giese NA et al. Pancreatic islet and
stellate cells are the main sources of endocrine gland-derived vascular endothelial
growth factor/prokineticin-1 in pancreatic cancer. Pancreatology 2009;9:165 172.
Zhong C, Qu X, Tan M, Meng YG, Ferrara N. Characterization and regulation of Bv8
in human blood cells. Clin Cancer Res 2009;15:2675 2684.
Pasquali D, Santoro A, Bufo P, Conzo G, Deery WJ, Renzullo A et al. Upregulation of
endocrine gland-derived vascular endothelial growth factor in papillary thyroid
cancers displaying infiltrative patterns, lymph node metastases, and BRAF mutation.
Thyroid 2011;21:391399.
Herr MM, Fries KM, Upton LG, Edsberg LE. Potential biomarkers of temporomandibular joint disorders. J Oral Maxillofac Surg 2011;69:4147.

Downloaded from by guest on March 26, 2015

61. Sitara D, Razzaque MS, Hesse M, Yoganathan S, Taguchi T, Erben RG et al. Homozygous ablation of fibroblast growth factor-23 results in hyperphosphatemia and
impaired skeletogenesis, and reverses hypophosphatemia in Phex-deficient mice.
Matrix Biol 2004;23:421 432.
62. Wessels A, Perez-Pomares JM. The epicardium and epicardially derived cells (EPDCs)
as cardiac stem cells. Anat Rec A Discov Mol Cell Evol Biol 2004;276:43 57.
63. Kruithof BP, van Wijk B, Somi S, Kruithof-de Julio M, Perez Pomares JM, Weesie F
et al. BMP and FGF regulate the differentiation of multipotential pericardial mesoderm
into the myocardial or epicardial lineage. Dev Biol 2006;295:507 522.
64. Smart N, Risebro CA, Melville AA, Moses K, Schwartz RJ, Chien KR et al. Thymosin
b4 induces adult epicardial progenitor mobilization and neovascularization. Nature
2007;445:177 182.
65. Pombal MA, Carmona R, Megas M, Ruiz A, Perez-Pomares JM, Munoz-Chapuli R. Epicardial development in lamprey supports an evolutionary origin of the vertebrate
epicardium from an ancestral pronephric external glomerulus. Evol Dev 2008;10:
210 216.
66. Simrick S, Masse K, Jones EA. Developmental expression of Pod 1 in Xenopus laevis. Int
J Dev Biol 2005;49:59 63.
67. Ferrara N, Frantz G, LeCouter J, Dillard-Telm L, Pham T, Draksharapu A et al. Differential expression of the angiogenic factor genes vascular endothelial growth factor
(VEGF) and endocrine gland-derived VEGF in normal and polycystic human
ovaries. Am J Pathol 2003;162:1881 1893.
68. Blowers E, Hall K. Adverse events in bevacizumab and chemotherapy: patient
management. Br J Nurs 2009;18:424 428.
69. Izzedine H. Angiogenesis inhibitor therapies: focus on hypertension and kidney
toxicity. [Article in French] Bull Cancer 2007;94:981 986.
70. Kappers MH, van Esch JH, Sleijfer S, Danser AH, van den Meiracker AH. Cardiovascular and renal toxicity during angiogenesis inhibition: clinical and mechanistic aspects.
J Hypertens 2009;27:2297 2309.
71. Hamada N, Kuwano K, Yamada M, Hagimoto N, Hiasa K, Egashira K et al. Antivascular endothelial growth factor gene therapy attenuates lung injury and fibrosis
in mice. J Immunol 2005;175:1224 1231.
72. Kang DH, Hughes J, Mazzali M, Schreiner GF, Johnson RJ. Impaired angiogenesis in the
remnant kidney model: II. Vascular endothelial growth factor administration reduces
renal fibrosis and stabilizes renal function. J Am Soc Nephrol 2001;12:1448 1457.
73. Chu TF, Rupnick MA, Kerkela R, Dallabrida SM, Zurakowski D, Nguyen L et al. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet 2007; 370:
2011 2019.
74. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer
2008;8:592 603.
75. Lindenmeyer MT, Kretzler M, Boucherot A, Berra S, Yasuda Y, Henger A et al.
Interstitial vascular rarefaction and reduced VEGF-A expression in human diabetic
nephropathy. J Am Soc Nephrol 2007;18:1765 1776.
76. Battersby S, Critchley HO, Morgan K, Millar RP, Jabbour HN. Expression and regulation of the prokineticins (endocrine gland-derived vascular endothelial growth

M. Boulberdaa et al.

You might also like