You are on page 1of 14

Coexpression of ecto-5′-nucleotidase/CD73 with

specific NTPDases differentially regulates adenosine


formation in the rat liver
Michel Fausther, Joanna Lecka, Elwy Soliman, Gilles Kauffenstein, Julie Pelletier,
Nina Sheung, Jonathan A. Dranoff and Jean Sévigny
Am J Physiol Gastrointest Liver Physiol 302:G447-G459, 2012. First published 1 December 2011;
doi:10.1152/ajpgi.00165.2011

You might find this additional info useful...

This article cites 57 articles, 16 of which can be accessed free at:


http://ajpgi.physiology.org/content/302/4/G447.full.html#ref-list-1

Updated information and services including high resolution figures, can be found at:
http://ajpgi.physiology.org/content/302/4/G447.full.html

Additional material and information about AJP - Gastrointestinal and Liver Physiology can be found at:
http://www.the-aps.org/publications/ajpgi

Downloaded from ajpgi.physiology.org on March 1, 2012


This infomation is current as of March 1, 2012.

AJP - Gastrointestinal and Liver Physiology publishes original articles pertaining to all aspects of research involving normal or
abnormal function of the gastrointestinal tract, hepatobiliary system, and pancreas. It is published 12 times a year (monthly) by the
American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2012 by the American Physiological
Society. ISSN: 0193-1857, ESSN: 1522-1547. Visit our website at http://www.the-aps.org/.
Am J Physiol Gastrointest Liver Physiol 302: G447–G459, 2012.
First published December 1, 2011; doi:10.1152/ajpgi.00165.2011.

Coexpression of ecto-5=-nucleotidase/CD73 with specific NTPDases


differentially regulates adenosine formation in the rat liver
Michel Fausther,1,2,3 Joanna Lecka,1 Elwy Soliman,2 Gilles Kauffenstein,1 Julie Pelletier,1 Nina Sheung,2
Jonathan A. Dranoff,2,3 and Jean Sévigny1
1
Centre de recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec (pavillon CHUL),
Québec and Département de microbiologie-infectiologie et d’immunologie, Faculté de médecine, Université Laval, Québec,
QC, Canada; 2Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and 3Division of
Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, University of Arkansas for Medical
Sciences, Little Rock, Arkansas
Submitted 27 April 2011; accepted in final form 29 November 2011

Fausther M, Lecka J, Soliman E, Kauffenstein G, Pelletier J, secretion, and scar formation (5, 19, 43, 44). To regulate
Sheung N, Dranoff JA, Sévigny J. Coexpression of ecto-5=-nucle- extracellular nucleoside and nucleotide signaling and maintain
otidase/CD73 with specific NTPDases differentially regulates adeno- tissue homeostasis, liver cells express various ectonucleoti-
sine formation in the rat liver. Am J Physiol Gastrointest Liver Phys- dases at their surface, including members of the ecto-nucleo-
iol 302: G447–G459, 2012. First published December 1, 2011;
side triphosphate diphosphohydrolase (NTPDase), ecto-nucle-

Downloaded from ajpgi.physiology.org on March 1, 2012


doi:10.1152/ajpgi.00165.2011.—Ectonucleotidases modulate puriner-
gic signaling by hydrolyzing ATP to adenosine. Here we characterized otide pyrophosphatase-phosphodiesterase, and alkaline phos-
the impact of the cellular distribution of hepatic ectonucleotidases, phatase families, as well as ecto-5=-nucleotidase/CD73 (5, 19,
namely nucleoside triphosphate diphosphohydrolase (NTPDase)1/CD39, 44). These ectoenzymes are thought to act in concert to
NTPDase2/CD39L1, NTPDase8, and ecto-5=-nucleotidase/CD73, and of sequentially dephosphorylate extracellular nucleotides and
their specific biochemical properties, on the levels of P1 and P2 receptor locally regulate their concentration in hepatic extracellular
agonists, with an emphasis on adenosine-producing CD73. Immunostain- fluids (55).
ing and enzyme histochemistry showed that the distribution of CD73 At physiological pH, plasma membrane-bound NTPDases,
(protein and AMPase activity) overlaps partially with those of namely NTPDase1/CD39, NTPDase2/CD39L1, and NTPDase8,
NTPDase1, -2, and -8 (protein levels and ATPase and ADPase activ- represent the major liver ectonucleotidase activities (18,
ities) in normal rat liver. CD73 is expressed in fibroblastic cells
located underneath vascular endothelial cells and smooth muscle cells,
45). NTPDases hydrolyze various nucleoside tri- and di-
which both express NTPDase1, in portal spaces in a distinct fibroblast phosphates (e.g., ATP and ADP) with distinct affinities and
population next to NTPDase2-positive portal fibroblasts, and in bile abilities. NTPDase1 hydrolyzes ATP and ADP equally well
canaliculi, together with NTPDase8. In fibrotic rat livers, CD73 whereas NTPDase2 preferentially breaks down ATP over
protein expression and activity are redistributed but still overlap with ADP, and NTPDase8 acts as a functional intermediate
the NTPDases mentioned. The ability of the observed combinations of between NTPDase1 and NTPDase2 (18, 26, 31). In the liver,
ectonucleotidases to generate adenosine over time was evaluated by NTPDases are expressed in a cell-specific manner: NTPDase1 is
reverse-phase HPLC with the recombinant rat enzymes at high “in- expressed by vascular endothelial cells and smooth muscle
flammatory” (500 ␮M) and low “physiological” (1 ␮M) ATP con- cells (18, 45), NTPDase2 by cells in the vascular external
centrations. Overall, ATP was rapidly converted to adenosine by the layer and by portal fibroblasts (14, 46), whereas NTPDase8
NTPDase1⫹CD73 combination, but not by the NTPDase2⫹CD73
combination. In the presence of NTPDase8 and CD73, ATP was is restricted to the canalicular membrane domain of hepa-
sequentially dephosphorylated to the CD73 inhibitor ADP, and then to tocytes (18).
AMP, thus resulting in a delayed formation of adenosine. In conclu- The monophosphonucleosides (e.g., AMP) generated by
sion, the specific cellular cocompartmentalization of CD73 with NTPDase activity are ultimately converted into the correspond-
hepatic NTPDases is not redundant and may lead to the differential ing nucleosides (e.g., adenosine) by ecto-5=-nucleotidase (12,
activation of P1 and P2 receptors, under normal and fibrotic condi- 27, 49). Interestingly, the NTPDase substrates ATP and ADP
tions. are natural inhibitors of the monophosphatase activity of ecto-
P1 receptors; P2 receptors; ATP; fibrosis; carbon tetrachloride intox- 5=-nucleotidase (39). In the liver, ecto-5=-nucleotidase protein
ication; nucleoside triphosphate diphosphohydrolases expression has been associated with the canalicular domain of
hepatocyte plasma membrane and undefined cellular elements
of the portal space (2, 47).
VIA THE ACTIVATION OF SPECIFIC plasma membrane P1 and P2 Because of their complementary biochemical properties,
receptors, extracellular nucleosides and nucleotides modulate NTPDases and ecto-5=-nucleotidase represent central elements
various physiological functions in virtually all tissues, includ- of liver purinergic signaling. However, the potential influence
ing blood vessels, heart, lungs, brain, smooth and skeletal of the coordinated action of NTPDases and ecto-5=-nucleoti-
muscle, etc. (9). In the liver, these signaling agents regulate key dase on local hepatic purinergic signaling has yet to be estab-
cellular pathways such as glucose metabolism, cell cycle pro- lished. In the present study, we have characterized the impact
gression, hepatic regeneration, cell volume regulation, ionic of the cellular distribution of these hepatic ectonucleotidases
on the local levels of P1 and P2 receptor agonists. Here we
Address for reprint requests and other correspondence: J. Sévigny, Centre de
show that ecto-5=-nucleotidase is coexpressed with NTPDase1,
Recherche en Rhumatologie et Immunologie, 2705 boul. Laurier, local T1-49, -2, and -8 in distinct liver compartments in normal and fibrotic
G1V 4G2 Québec, QC, Canada (e-mail: Jean.Sevigny@crchul.ulaval.ca). rat liver. These specific associations of ecto-5=-nucleotidase
http://www.ajpgi.org 0193-1857/12 Copyright © 2012 the American Physiological Society G447
G448 FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES

with each hepatic NTPDase have the ability to differentially 7.4) and the products of ATP hydrolysis were determined by reverse
regulate the signaling pathways modulated by extracellular phase HPLC, as previously described (31).
nucleotides and their metabolites in the liver. For assays at low ATP concentration (1 ␮M), ectonucleotidase-
overexpressing crude protein extracts were prepared with an ATPase
(for NTPDase) and AMPase activity (for ecto-5=-nucleotidase) suffi-
EXPERIMENTAL PROCEDURES
cient to convert ⬃40% of the initial substrate within 5 min, as
Materials determined by HPLC. Enzymatic reactions were performed at 37°C
for 30 min in a modified Krebs buffer (114 mM NaCl, 4.7 mM KCl,
Culture media were obtained from Invitrogen (Burlington, ON, 1.2 mM KH2PO4, 1.16 mM MgSO4, 2.5 mM CaCl2, 2.5 MgCl2, 25.5
Canada). Adenosine 5=-(␣,␤-methylene)diphosphate (␣,␤-methylene- mM NaHCO3, 20 mM HEPES, pH 7.4). A modified Krebs buffer
ADP), 2-chloroacetaldehyde, 3,3=-diaminobenzidine (DAB), 1,4- was used here instead of Ringer buffer because of interference with
diaza-bicyclo[2.2.2]octane (DABCO), nucleotides, levamisole, tet- fluorescence detection observed with the latter. Briefly, 150-␮l
rabutylammonium hydrogen sulfate were provided by Sigma-Aldrich aliquots of reaction mixture were collected after various time
(Oakville, ON, Canada). The 4=,6-diamidino-2-phenylindole (DAPI) intervals and transferred to an equal volume of ice-cold 5%
dye was purchased from Molecular Probes (Eugene, OR) and aqueous (wt/vol) trichloroacetic acid (TCA) for deproteinization. The sam-
hematoxylin from Biomeda (Foster City, CA). Mouse monoclonal ples were centrifuged at 10,000 g for 3 min at 4°C and the
antibody to rat CD31/PECAM-1 (clone TLD-3A12) was purchased supernatants were subjected to lipid extraction with diethyl ether
from BD Pharmingen (Mississauga, ON, Canada), to rat CD68 (clone (3:1, vol/vol; 5 cycles). The resulting samples were subsequently
ED-1) from AbD Serotec (Raleigh, NC), and those to mouse/rat transformed to etheno(ε)-derivatives, according to a modified der-
␣-smooth muscle actin (SMA; clone 1A4) and rat vimentin (clone V9) ivatization protocol by Levitt et al. (34, 36). This additional step
from Sigma-Aldrich. The antibodies to NTPDases and ecto-5=-nucle- allowed the determination of 50-fold lower concentrations of

Downloaded from ajpgi.physiology.org on March 1, 2012


otidase can be obtained from ectonucleotidases-ab.com (Québec, QC, adenylated species by fluorescence detection.
Canada). In a representative derivatization assay, a 200-␮l aliquot of the
extracted sample was incubated for 60 min at 70°C in the presence
Plasmids of 1 M chloroacetaldehyde and 25 mM Na2HPO4, pH 4.0, in a final
volume of 250 ␮l. The resulting fluorescent 1,N6-ethenoadenine
The plasmids encoding rat NTPDase1 (NM_022587) (26), derivatives (ε-ATP, ε-ADP, ε-AMP, and ε-adenosine) were sepa-
NTPDase2 (NM_172030) (30), NTPDase8 (AY536920) (18), and rated by reverse-phase HPLC as previously described (31), where
ecto-5=-nucleotidase/CD73 (NM_021576) (8) were used for antiserum the mobile phase used contained 25 mM tetrabutylammonium
generation and for cell transfection, as described below. hydrogen sulfate and 100 mM KH2PO4/K2HPO4, pH 7.0, in 10%
(vol/vol) MeOH. All experiments were performed in triplicate.
Animals and Antibody Production Samples to which crude protein extracts were added after reaction
Sprague-Dawley rats, Hartley guinea pigs and New Zealand rabbits had been stopped (i.e., after TCA addition) were used as controls
were obtained from Charles River Laboratories (St-Constant, QC, (baseline levels). Separation and detection of ethenylated deriva-
Canada) or Harlan Sprague Dawley (Indianapolis, IN). All procedures tives were performed with an automated Waters and Beckman
were approved by the Canadian Council on Animal Care and by the HPLC apparatus equipped with an RF-10AXL Shimadzu fluores-
Laval University Animal Ethics Advisory Committee and the Yale cence detector (␭exc ⫽ 307 nm; ␭em ⫽ 410 nm). Quantification and
Institutional Animal Care and Use Committee. Genetic immunization identification of the fluorescent adenylated derivatives were carried
was carried out by direct injection of a plasmid encoding rat ecto-5=- out by comparison with the corresponding ε-purine standards.
nucleotidase (8), as previously described with other antigens (18), in
Immunoblotting
Hartley guinea pigs and New Zealand rabbits.
Chronic liver fibrosis was induced in rats by carbon tetrachloride Protein preparation, SDS-PAGE fractionation, and electroblotting
(CCl4) intoxication. For CCl4 treatment, male rats were provided with were performed as previously described (18). Following incubation
drinking water containing 1% phenobarbital starting 2 wk before and with primary antibodies, proteins of interest were visualized with the
until the end of CCl4 treatment, to upregulate cytochrome P-450 appropriate horseradish peroxidase-conjugated secondary antibodies
enzyme levels (24). Rats were injected with CCl4 (0.1 ml sc, CCl4 (anti-rabbit IgG, Amersham Biosciences, Boston, MA; anti-guinea pig
diluted 1:6 in olive oil) three times per week for 6 – 8 wk. For control IgG, Santa Cruz Biotechnology, Santa Cruz, CA) and the Chemilu-
experiments, rats were injected in identical fashion with olive oil minescent Reagent Plus (Perkin-Elmer, Boston, MA), according to the
alone. Livers were harvested at 6 – 8 wk and processed for enzyme manufacturer’s recommendations.
histochemistry as described below.
Immunofluorescence, Immunocytochemistry, and
Cell Transfection and Ectonucleotidase Activity Assays Enzyme Histochemistry
Transfection of COS-7 cells using plasmids encoding either recom- COS-7 cells (105/coverslip) or 6-␮m sections of snap-frozen rat
binant NTPDase1, -2, -8, or ecto-5=-nucleotidase and preparation of liver specimens were fixed with cold acetone:phosphate-buffered
crude protein extracts were carried out as previously described (31). formalin (9:1). For immunofluorescence, fixed sections were incu-
The COS-7 cell line was used because it exhibits very low basal levels bated with the following polyclonal antibodies against rat enzymes:
of nucleotide- and nucleoside-metabolizing enzyme activities (Refs. anti-NTPDase1 (rN1– 6L) (18), anti-NTPDase2 (BZ3– 4F) (14), anti-
29, 51 and data not shown). NTPDase8 (rN8 – 8C) (18), and anti-CD73 (either r5=NT-4C or r5=NT-
For assays at high ATP concentration (500 ␮M), ectonucleotidase- 9L), or corresponding preimmune serum samples. The appropriate
overexpressing crude protein extracts were added to obtain an enzy- Alexa Fluor-conjugated secondary antibodies (Molecular Probes)
matic activity of 24 nmol Pi/min with ATP and AMP as substrate for were then applied as previously described (18). In one set of double
NTPDases and ecto-5=-nucleotidase, respectively, as determined by immunostaining experiments, rabbit rN1– 6L and BZ3– 4F antibodies
the malachite green procedure according to Baykov et al. (3, 32). The were each used in combination with guinea pig r5=NT-4C serum, and
enzymatic reactions were performed at 37°C for 150 min in Ringer the guinea pig rN8 – 8C antibody was used with rabbit r5=NT-9L serum.
buffer (120 mM NaCl, 5 mM KCl, 2.5 mM CaCl2, 2.5 mM MgCl2, 1.2 In the other set of experiments, mouse monoclonal antibodies against
mM MgSO4, 25 mM NaHCO3, 10 mM dextrose, 80 mM Tris·HCl, pH rat CD31/PECAM-1 (clone TLD-3A12), rat ␣-SMA (clone 1A4) and

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES G449
rat vimentin (clone V9) were each used in combination with the rabbit Flow Cytometry and Isolation of Rat Liver Cell Populations
r5=NT-9L serum.
For immunocytochemistry, fixed cells were incubated with the Hepatocytes, hepatic stellate cells, portal fibroblasts and nonparen-
polyclonal CD73 antiserum (r5=NT-4C and r5=NT-9L) and the appro- chymal cell preparations were isolated from fresh rat liver tissue as
priate biotinylated secondary antibodies (Jackson ImmunoResearch previously described (14, 16). Fixed primary isolated cells were
Laboratories, Westgrove, PA) before addition of Vectastain Elite labeled by indirect immunofluorescence after incubation with a rabbit
ABC reagent (Vector Laboratories, Burlington, ON, Canada). All polyclonal serum to rat CD73 (r5=NT-9L) followed by Alexa Fluor
antibodies were used at a 1:1,000 working dilution, except for r5=NT- 488-conjugated polyclonal anti-rabbit IgG antibody (Molecular
9L, which was used at a final concentration of 1:2,000. Endogenous Probes). Cell-surface expression of CD73 protein was assessed by
peroxidase was quenched by incubating the fixed cells with 0.15% fluorescence-activated cell sorting, using a FACSCalibur (BD Biosci-
(vol/vol) H2O2 (in PBS). Endogenous avidin and biotin were blocked ences, Rockville, MD) sorter and Flowjo software Version 8.7 (Tree-
using the Avidin/Biotin Blocking Kit (Vector Laboratories). The Star, Ashland, OR).
peroxidase reaction was performed in a solution containing the DAB
RESULTS
substrate (0.4 mg/ml) and 0.03% (vol/vol) H2O2 in PBS, and stopped
with extensive water rinsing. Although the hepatic expression of NTPDase1, -2, and -8
For enzyme histochemistry, ectonucleotidase activities were local- has previously been reported (14, 18, 45), the cellular local-
ized by using the Wachstein/Meisel lead phosphate method as mod-
ified by Braun et al. (see reference therein) (7). The enzymatic ization of ecto-5=-nucleotidase/CD73 had not been clearly
reaction was carried out by incubating liver sections with various established. Therefore, we first looked at the distribution of
nucleotides (200 ␮M) as substrates for 1 h. All experiments were ecto-5=-nucleotidase in relation to NTPDases, using enzyme
performed in the presence of levamisole (5 mM), an inhibitor of histochemical and immunohistochemical methods.

Downloaded from ajpgi.physiology.org on March 1, 2012


tissue-nonspecific alkaline phosphatase (TNAP). In a separate set
of experiments, ␣,␤-methylene-ADP (1 mM) was used as an inhibitor Histochemical Detection of Ectonucleotidase Activities in
of ecto-5=-nucleotidase. Control assays (not shown) were performed Normal and Fibrotic Rat Liver
in the absence of nucleotide.
Nuclei were counterstained with either DAPI (immunofluores- All enzyme histochemistry assays were performed in the
cence) or aqueous hematoxylin (immunocytochemistry and enzyme presence of the TNAP inhibitor levamisole (5 mM) to exclude
histochemistry). Mowiol 4-88 medium (supplemented with DABCO any contribution of this enzyme to the apparent catalytic
as antifade reagent for immunofluorescence only) was used as mount- activities measured with the nucleotide substrates. Neverthe-
ing medium. less, no difference in ectonucleotidase activity could be de-

Fig. 1. Histochemical detection of liver ecto-


nucleotidase activities. Ectonucleotidase activ-
ities were located in serial sections of a normal
rat liver, using various nucleotides (200 ␮M)
as substrates. All 3 structural components of a
classical hepatic triad are depicted, including
branches of hepatic artery (A), portal vein (V),
and intrahepatic bile duct (BD). ATPase and
ADPase catalytic products are mainly detected
in all vasculature (arterial ⬎ venous), in the
connective tissue associated with the portal
space, and in the bile canaliculi (BC) (top and
middle). The AMP hydrolysis pattern is located
in structures similar to those where ATPase and
ADPase activities are found, although no signal
is observed either in endothelium or in vascular
smooth muscle (arrows, bottom). No ectonucle-
otidase activity is detected in cholangiocytes for
any of the substrates tested (asterisks, right). All
enzyme histochemical assays were performed in
the presence of levamisole (5 mM) to inhibit
tissue-nonspecific alkaline phosphatase (TNAP)
activity. Scale bar, 10 ␮m.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


G450 FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES

tected when omitting levamisole from the reaction solution ATPase, ADPase, and AMPase activities were observed in
(data not shown), suggesting that TNAP activity was negligible fibrous septa surrounding hepatocyte nodules, in perivascular
under the experimental conditions used. In the rat liver, en- connective tissue areas, as well as in cells localized within
zyme histochemical assays using ATP as substrate revealed a hepatic sinusoids. Both ATPase and ADPase hydrolytic activ-
prominent staining activity in vascular endothelium and sub- ities were still observed along the hepatocyte plasma mem-
endothelium, in the connective tissue adjacent to the portal/ brane, with a more prominent but disrupted canalicular dis-
periportal space and surrounding central veins, in sinusoids, tribution, whereas AMPase activity was increased at all
and in bile canaliculi (Fig. 1; ATP). No nucleotide hydrolysis levels of the hepatocyte membrane. These observations,
was detected in ductular epithelium. Hepatic ADPase activity although qualitative, indicate that levels and distribution of
displayed a distribution pattern similar to ATPase activity, ATPase, ADPase, and AMPase activities are regulated dur-
albeit with a slightly weaker staining intensity (Fig. 1; ADP). ing hepatic fibrosis and suggest their involvement in the
When AMP was applied as substrate, catalytic products were inflammatory liver response. This is in agreement with
mainly detected in the subendothelium, in the connective tissue previous studies showing that transcriptional regulation of
associated with the perivascular and periductular areas, and in ectonucleotidase gene expression occurs during liver in-
the bile canaliculi. Note that ductular epithelium, vascular endo- flammation and fibrosis (15, 23, 40, 53).
thelium, as well as smooth muscle were devoid of AMPase activity
(Fig. 1; AMP). Lack of endothelial AMPase activity was Generation of Polyclonal Antibodies to Rat
observed in blood vessels of all types and calibers. When Ecto-5=-Nucleotidase/CD73 and Specificity
␣,␤-methylene-ADP (1 mM) was added to specifically inhibit

Downloaded from ajpgi.physiology.org on March 1, 2012


ecto-5=-nucleotidase activity, overall AMP hydrolysis was re- To determine the cellular distribution of ecto-5=-nucleoti-
duced in all regions, suggesting that this enzyme contributes to dase and the various NTPDases in the liver, we used a panel of
most, if not all, AMPase activity present in the rat liver at polyclonal antibodies developed in our laboratory. The speci-
physiological pH (Fig. 2). In support of these findings, enzyme ficity of polyclonal sera to NTPDase1 (rN1– 6L), NTPDase2
histochemical assays performed with other ecto-5=-nucleoti- (BZ3– 4F), and NTPDase8 (rN8 – 8C) has previously been val-
dase substrates, namely CMP, GMP, and IMP, showed similar idated (14, 18). Guinea pig and rabbit polyclonal antibodies
patterns of activity (not shown). directed against rat ecto-5=-nucleotidase have been generated
When enzyme histochemistry assays were performed in and their specificity tested by the following immunological tech-
livers from rats subjected to CCl4 intoxication (Fig. 3), hepatic niques. By immunocytochemistry, positive dark brown staining
fibrosis induction was associated with obvious alterations of could be seen only with ecto-5=-nucleotidase-transfected COS-7
ectonucleotidase activity distribution. Strong signals for cells incubated in presence of either r5=NT-4C guinea pig serum or

Fig. 2. Histochemical detection of liver ecto-5=-nucleotidase activity. Ecto-5=-nucleotidase activity was located in normal rat liver sections with AMP (200 ␮M)
as substrate. Top: prominent signals for hepatic AMPase activity in the connective tissue associated with perivascular areas (arrows), and in bile canaliculi
(asterisks). Bottom: ecto-5=-nucleotidase inhibitor ␣,␤-methylene-ADP (␣␤-MeADP; 1 mM) reduces AMPase activity in all structures (see arrows and asterisks).
All enzyme histochemical assays were performed in the presence of the TNAP inhibitor levamisole (5 mM). Scale bar, 20 ␮m.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES G451

Fig. 3. Histochemical detection of liver ectonucleotidase activ-


ities in CCl4-induced hepatic fibrosis model. Ectonucleotidase
activities were located in liver sections from vehicle (control)
and CCl4-treated rats, using ATP, ADP, or AMP (200 ␮M) as
substrate. All 3 structural components of a classical hepatic
triad are depicted: branches of hepatic artery, portal vein, and
the intrahepatic bile duct. ATPase, ADPase, and AMPase
activities in control animals followed a distribution pattern

Downloaded from ajpgi.physiology.org on March 1, 2012


similar to that described in Fig. 1. In contrast, in fibrotic
animals, overlapping signals for ATP, ADP, and AMP hydro-
lytic products were detected in fibrous bands surrounding
hepatic nodules. Moreover, staining for ATPase and ADPase
activities was observed in hepatic sinusoids and hepatocyte
basolateral membrane, and more prominently, although in a
diffuse manner, in bile canaliculi. All enzyme histochemical
assays were performed in the presence of the TNAP activity
inhibitor levamisole (5 mM). CV, central vein; FS, fibrous
septum. Scale bar, 10 ␮m.

r5=NT-9L rabbit serum, respectively (Fig. 4, A and B; serum). The CD68-positive; Fig. 5A and data not shown). As seen with
same antisera detected a strong protein band with a molecular BZ3– 4F serum, NTPDase2 was detected in the subendothelial
weight of ⬃66 kDa by immunoblotting in the lane corresponding basement membrane cells, in portal fibroblasts, and in peri-
to the crude protein extracts from ecto-5=-nucleotidase-transfected ductular “bundle-shaped” structures, in the connective tissue of
cells. No protein was detected in the lanes with crude protein portal space area, likely representing intrahepatic nerves (Fig.
extracts from nontransfected cells, confirming the specificity of 5B). Staining with the rN8 – 8C polyclonal antibody showed
both types of polyclonal antibodies (Fig. 4, A and B; immunoblot). that NTPDase8 expression was restricted to bile canaliculi
Furthermore, both r5=NT-4C and r5=NT-9L antibodies displayed (Fig. 4C). Both r5=NT-4C and r5=NT-9L polyclonal sera re-
similar distribution patterns for hepatic ecto-5=-nucleotidase, as vealed strong ecto-5=-nucleotidase expression in the connective
determined by immunohistochemistry and immunofluorescence tissue associated with the perivascular and periductular areas,
(see next section; data not shown). as well as in bile canaliculi, with a faint immunostaining in the
basal membrane of hepatocytes (Fig. 4; ECTO-5=NT, all pan-
Immunolocalization of NTPDases and Ecto-5=-Nucleotidase
els). Interestingly, ecto-5=-nucleotidase expression was absent
in Normal and Fibrotic Rat Liver
from the vascular endothelium (Fig. 5; ECTO-5=NT, all pan-
Immunofluorescence labeling in the rat liver with the polyclonal els). Double immunofluorescence staining showed that al-
antibody rN1–6L showed prominent staining for NTPDase1 in vas- though ecto-5=-nucleotidase did not strictly colocalize with
cular endothelium and smooth muscle cells as well as in liver NTPDase1, both proteins were located in the same structures in
resident macrophages, namely Kupffer cells (detected as adjacent cells, such as NTPDase1-positive Kupffer cells pres-

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


G452 FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES

Fig. 4. Specificity of the polyclonal antibodies to


rat ecto-5=-nucleotidase. Immunocytochemistry
was performed with intact nontransfected (insets)
or transfected COS-7 cells with an expression
vector encoding rat ecto-5=-nucleotidase. Cells
were incubated with either preimmune serum or
guinea pig (r5=NT-4C; top) or rabbit (r5=NT-9L;
bottom) polyclonal antibody to ecto-5=-nucleoti-
dase. Immunoreactivity is present only in trans-
fected cells incubated with either antibody. Scale
bar, 20 ␮m. Immunoblot analysis of lysates from
nontransfected COS-7 cells (ctrl) next to lysates
from COS-7 cells transfected with the ecto-5=-
nucleotidase expression vector (CD73) shows a
single band of ⬃66 kDa found exclusively in
lysates from transfected cells, again confirming
the specificity of both guinea pig r5=NT-4C and
rabbit r5=NT-9L sera. A: guinea pig antibody

Downloaded from ajpgi.physiology.org on March 1, 2012


(r5=NT-4C; right) or its preimmune serum (left).
B: rabbit antibody (r5=NT-9L; right) or its preim-
mune serum (left).

ent in perivascular and/or periductular connective tissue, and hepatocyte bile canaliculi, appeared rather reduced and diffuse
on the basolateral surface of the endothelial cell layer in close in these structures compared with control liver. Finally, in-
contact with the perivascular extracellular matrix (Fig. 5A, creased ecto-5=-nucleotidase expression was observed in fi-
merged). Likewise, although ecto-5=-nucleotidase did not co- brotic septa and sinusoidal spaces between hepatocytes. Also,
localize with NTPDase2, both enzymes were expressed in the its canalicular distribution at the hepatocyte membrane level
same vicinity in the perivascular and periductular connective was diffuse and less prominent relatively to control liver.
tissue areas (Fig. 5B, merged). By contrast, NTPDase8 and Again, as noted in normal rat liver, the distribution profiles of
ecto-5=-nucleotidase colocalized in bile canaliculi (Fig. 5C, NTPDase1, -2, -8, and ecto-5=-nucleotidase enzymes paral-
merged). leled those described for ATPase, ADPase (NTPDases), and
We further investigated ecto-5=-nucleotidase expression in AMPase (ecto-5=-nucleotidase) activities in fibrotic livers.
the rat liver using the following cell markers: cell adhesion Taken together, these data show that the histochemical local-
molecule PECAM-1 for vascular endothelium, ␣-SMA for ization of ATPase and ADPase activities correlates with the
smooth muscle cells and myofibroblasts, and vimentin for combined distribution of NTPDase1, -2, and -8, as detected by
fibroblasts and other liver mesenchymal cells (42). Ecto-5=- immunohistochemistry, whereas the AMPase activity corresponds
nucleotidase labeling was clearly distinct from that observed to the immunohistological expression pattern of ecto-5=-nucleoti-
for PECAM-1 and ␣-SMA, but partly overlapped with vimen- dase in normal and fibrotic rat livers. In addition, our results
tin staining in the perivascular and periductular areas (Fig. 6; suggest that ecto-5=-nucleotidase is expressed by a fibroblastic
merged, all panels). We further confirmed the expression of population that is devoid of NTPDase activity in normal rat liver.
ecto-5=-nucleotidase in primary liver cell preparations by flu-
orescence-activated cell sorting analysis. These data demon- HPLC Analysis of ATP Hydrolysis by Different Hepatic
strate that in normal rat liver, ecto-5=-nucleotidase is expressed NTPDases in Combination with Ecto-5=-Nucleotidase
by NTPDase8-positive hepatocytes and a nonparenchymal cell
type that is distinct from hepatic stellate cells and NTPDase2- Based on the localization determined as above for NTPDase
positive portal fibroblasts (Fig. 7). and ecto-5=-nucleotidase activities and protein expression, we
We also investigated the distribution of ectonucleotidases evaluated how the net biochemical activity resulting from the pairing
NTPDase1, -2, -8, and ecto-5=-nucleotidase in specimens from of ecto-5=-nucleotidase with either NTPDase1, NTPDase2, or
CCl4-induced fibrosis by immunohistochemistry (Fig. 8). Pos- NTPDase8 might affect the generation of nucleotide and nu-
itive staining for NTPDase1 expression could be observed in cleoside species in the liver. As described in EXPERIMENTAL
several proliferating vascular structures and (likely) inflamma- PROCEDURES, ATP hydrolysis assays were performed at a high,
tory cells in liver fibrotic areas and intersinusoidal spaces. enzyme-saturating substrate concentration (500 ␮M) such as
NTPDase2 distribution was noticeably reorganized and present found under pathological conditions such as inflammation or
in fibrous septa surrounding hepatic nodules, as previously hepatic failure (21, 25, 55), as well as at low more physiolog-
described (15). NTPDase8 expression, while still restricted to ical substrate concentration (1 ␮M) (10, 33). Aliquots of the

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES G453

Downloaded from ajpgi.physiology.org on March 1, 2012


Fig. 5. Comparative distribution of liver nucleoside triphosphate diphosphohydrolases (NTPDases) and ecto-5=-nucleotidase (ECTO-5=NT). Double immuno-
fluorescence labeling of NTPDases and ecto-5=-nucleotidase was performed in serial sections of rat liver. A: NTPDase1 staining is associated with the liver
vascular network and Kupffer cells. B: NTPDase2 labeling is detected in the connective tissue of portal and septal spaces and perivascular areas. C: NTPDase8
signal is present exclusively in bile canaliculi. A–C: ecto-5=-nucleotidase immunostaining is observed in the connective tissue found in portal space or
perivascular areas as well as in bile canaliculi. Double staining of NTPDase1 and ecto-5=-nucleotidase reveals that both ectoenzymes are expressed by different
but adjacent cell populations in the vasculature, hepatic sinusoids, and hepatic extracellular matrix (A, MERGED). Similarly, NTPDase2 and ecto-5=-nucleotidase
proteins are both present at the surface of distinct cell types of hepatic connective tissue (B, MERGED). Both NTPDase8 and ecto-5=-nucleotidase colocalize
in bile canaliculi (C, MERGED). Scale bar, 10 ␮m except for A, bottom, where scale bar is 20 ␮m. DAPI, 4’,6-diamidino-2-phenylindole.

reaction mixtures were collected at different time points, and AMP was rapidly produced, the formation of adenosine by
nucleotide and nucleoside contents were evaluated by HPLC. ecto-5=-nucleotidase appeared to be slow during the first hour,
The time course of ATP hydrolysis and time-dependent gen- likely because of the inhibitory effect of ATP and ADP
eration of adenosine were analyzed for the different pairs of rat transiently present in the medium at that early stage. When
ectonucleotidases observed above for each liver structure. both ATP and ADP concentrations decreased afterward, aden-
High ATP concentration (500 ␮M). In the presence of osine formation increased until full conversion of AMP to
NTPDase1 and ecto-5=-nucleotidase, ATP was hydrolyzed rap- adenosine. When ATP was incubated with NTPDase2 and
idly to adenosine, with transient increases of ADP and AMP ecto-5=-nucleotidase, it was readily converted into ADP, but
concentrations (Fig. 9, NTPDase1 and CD73, high). Although very poorly into AMP, as predicted by the fact that ADP is a

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


G454 FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES

Downloaded from ajpgi.physiology.org on March 1, 2012


Fig. 6. Comparative distribution of liver ecto-5=-nucleotidase with cell markers. Double immunofluorescence labeling of ecto-5=-nucleotidase and cell markers
for vascular endothelial cells [platelet/endothelial cell adhesion molecule-1 (PECAM-1)], myofibroblastic cells (␣-smooth muscle actin), and nonmyofibroblastic
cells (vimentin) was performed in rat liver sections. Although ecto-5=-nucleotidase immunostaining is clearly distinct from that of PECAM-1 and ␣-SMA, it
partially overlaps with vimentin staining. Scale bar, 10 ␮m.

poor substrate of NTPDase2 (Fig. 9, NTPDase2 and CD73, ⬃80% of ATP added was hydrolyzed to adenosine. The ADP
high). Because of the protracted accumulation of ADP in the generated upon ATP hydrolysis by the NTPDase2 plus ecto-
assay medium and the low abundance of AMP substrate for 5=-nucleotidase pair accumulated in the reaction medium, with
ecto-5=-nucleotidase, adenosine formation was accordingly little AMP production, and as a result, adenosine was produced
limited. Of note is the observation that under the conditions only in very small amounts (Fig. 9, NTPDase2 and CD73,
described, rat NTPDase2 was active only for 50 – 60 min. The low). The pattern of ATP hydrolysis by NTPDase8 plus ecto-
ATP hydrolysis profile obtained with the pairing of NTPDase8 5=-nucleotidase was very similar to the latter combination and
with ecto-5=-nucleotidase showed a rapid ADP accumulation, resulted in the continuous accumulation of ADP (Fig. 9,
peaking after 20 min, which was followed by the delayed NTPDase8 and CD73, low) with limited production of AMP
generation of AMP, which reached a plateau between 50 and and adenosine. In the presence of 1 ␮M ATP, the level of ATP
120 min (Fig. 9, NTPDase8 and CD73, high). These observa- and/or ADP produced was apparently too low to inhibit ecto-
tions concur with the fact that NTPDase8 activity is higher
5=-nucleotidase. In this condition, the rate of adenosine forma-
with ATP than with ADP. As a result, ecto-5=-nucleotidase
tion was solely dependent on the substrate (AMP) generated.
activity was transiently inhibited by the presence of ADP,
which delayed adenosine formation compared with the hydro- This contrasts with the data obtained at a high ATP concen-
lysis profile obtained with the NTPDase1 plus ecto-5=-nucle- tration (Fig. 9 NTPDase8 and CD73, high) where the “higher”
otidase pair. concentration of ADP generated was hampering the ability of
Low ATP concentration (1 ␮M). In the presence of the ecto-5=-nucleotidase to hydrolyze AMP to adenosine, in agree-
NTPDase1 and ecto-5=-nucleotidase pair, ATP was directly ment with our previous observations (35).
converted to AMP, ADP being rapidly hydrolyzed after it was Note that in these assays we did not detect any degradation
formed (Fig. 9, NTPDase1 and CD73, low). Therefore, aden- products of adenosine, indicating the lack of expression of
osine formation by ecto-5=-nucleotidase almost paralleled adenosine-metabolizing enzymes in COS-7 cells, in keeping
AMP appearance in the reaction mixture. After about 20 min, with the report of Tkacz et al. (51).

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES G455
activity (14, 18) in normal rat liver, different combinations of
NTPDase and ecto-5=-nucleotidase expression patterns were
found: 1) NTPDase1 (vascular endothelium, smooth muscle
cells, and Kupffer cells) and ecto-5=-nucleotidase, although not
actually expressed in the same cells, were collectively present
at the junction between the vascular tunica media and the
perivascular connective tissue or at the sinusoidal face of
hepatocyte membranes; 2) NTPDase2 (portal fibroblasts) and
ecto-5=-nucleotidase were also expressed by neighboring cells
located within the portal/periportal connective tissue; and fi-
nally 3) NTPDase8 and ecto-5=-nucleotidase were coexpressed
by hepatocytes in bile canaliculi. It is noteworthy to mention that
ecto-nucleotide pyrophosphatases/phosphodiesterases, which have
the ability to hydrolyze tri- and diphosphate nucleosides in
principle, are also expressed in the liver (48). However, their
biochemical properties, namely a lower substrate affinity than
Fig. 7. Flow cytometry analysis of ecto-5=-nucleotidase expression in liver NTPDases and optimal hydrolytic activity at alkaline rather
primary cell populations. Cell populations were prepared as described in
EXPERIMENTAL PROCEDURES. Ecto-5=-nucleotidase (CD73) was labeled using
than physiological pH, did not allow us to evaluate their
rabbit polyclonal r5=NT-9L serum. Isolated hepatocytes and nonparenchymal potential contribution to liver purinergic signaling in our con-

Downloaded from ajpgi.physiology.org on March 1, 2012


cells (NPCs) are positive for ecto-5=-nucleotidase staining whereas portal dition settings.
fibroblasts (PFs) and hepatic stellate cells (HSCs) are mostly negative. Induction of experimental liver fibrosis in CCl4-treated rats
was associated with changes in the distribution of all hepatic
DISCUSSION ectonucleotidase activities. Overall, ATPase, ADPase, and
AMPase activities were increased in liver fibrotic areas, in
In the present study, we demonstrate a correlation between sinusoidal spaces, and in fibrous septa in the periphery of
ecto-5=-nucleotidase/CD73 protein expression and AMPase hepatic nodules, a hallmark of CCl4-induced fibrosis. In the
activity in both normal and fibrotic rat livers. In normal rat same fibrotic livers, NTPDase1 protein was associated with
liver, both attributes of ecto-5=-nucleotidase were detected in proliferating blood vessels, NTPDase2 protein with the fibrous
the canalicular and sinusoidal membrane domains of hepato- septa surrounding hepatic nodules, and NTPDase8 protein with
cytes as well as in the connective tissue surrounding central structurally altered bile canaliculi of hepatocytes. Ecto-5=-
veins or associated with the portal and larger septal space nucleotidase protein expression was detected in both apical and
areas. Quite notably, ecto-5=-nucleotidase protein expression sinusoidal domains of hepatocyte plasma membrane as well as
and AMPase activity were absent from the cell surface of in fibrous septa. Again, as observed in normal rat liver, hepatic
hepatic vascular endothelium and bile duct epithelium in rat, as AMPase activity and expression of ecto-5=-nucleotidase over-
shown here, and in mouse (data not shown). The absence of lapped with the localization of hepatic ATPase and ADPase
ecto-5=-nucleotidase from murine endothelial cells contrasts activities as well as the expression of NTPDase1, -2, and -8.
with the vascular localization reported for its human ortholog We addressed the potential impact of this specific distribu-
(Ref. 1 and data not shown). Our examination of ecto-5=- tion of NTPDases and ecto-5=-nucleotidase on extracellular
nucleotidase distribution by double fluorescence immunostain- nucleotide levels by performing a biochemical ATP hydrolysis
ing with specific cell markers as well as flow cytometry assay in the presence of each pair formed by ecto-5=-
analysis of primary liver cells further indicates that ecto-5=- nucleotidase with either NTPDase1, -2, or -8. It is notewor-
nucleotidase is mainly expressed in hepatocytes and nonmyo- thy that the hydrolytic patterns observed in Fig. 9 are in
fibroblastic (nonparenchymal) liver cells that are likely distinct agreement with the reported kinetic properties of each
from portal fibroblasts and hepatic stellate cells. Importantly, NTPDase (31) and with the fact that ATP and ADP are
the facts that 1) levamisole, an inhibitor of alkaline phospha- physiological inhibitors of ecto-5=-nucleotidase (35). Indeed, at
tase activity, had no effect on the AMPase activity observed 500 ␮M ATP, a significant decrease in ATP and ADP concen-
(data not shown), 2) hydrolysis of AMP, CMP, GMP, and IMP tration was required to observe the effective production of
(Figs. 1 and 2 and data not shown) all matched ecto-5=- adenosine from ecto-5=-nucleotidase. As expected, adenosine
nucleotidase immunolocalization using two different antibod- generation was fastest in the presence of NTPDase1, which
ies (Figs. 4 and 5 and data not shown), and 3) this activity was rapidly hydrolyzed both ATP and ADP to the ecto-5=-nu-
decreased by ␣,␤-methylene-ADP, an ecto-5=-nucleotidase in- cleotidase substrate, AMP. In contrast, in the presence of
hibitor (Fig. 2) suggest that ecto-5=-nucleotidase is responsible NTPDase2, adenosine production was very low because of the
for most if not all nucleoside monophosphatase activity present very limited hydrolysis of ADP by this enzyme. The result
in the liver. Ecto-5=-nucleotidase therefore appears to be the obtained with NTPDase8 was somewhat intermediate between
enzyme responsible for the formation of extracellular adeno- those observed for NTPDase1 and -2 at 500 ␮M ATP, which
sine at physiological pH in the rat liver. This enzyme would correlates with a transient production of ADP by NTPDase8
therefore be expected to play a key role in the regulation of P1 (18, 31), with adenosine production being inhibited when ATP
receptor activation. and ADP levels exceed 50 –100 ␮M, even at high AMP levels
When we compared the distribution of ecto-5=-nucleotidase (200 ␮M). Interestingly, the situation was different for hydro-
to that of hepatic NTPDase1, -2, and -8 with respect to the lysis of low ATP concentration (1 ␮M), where ATP and ADP
same parameters, i.e., protein expression and ectonucleotidase levels were insufficient to inhibit ecto-5=-nucleotidase, and as

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


G456 FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES

Downloaded from ajpgi.physiology.org on March 1, 2012


Fig. 8. Comparative distribution of liver NTPDases and ecto-
5=-nucleotidase in CCl4-induced hepatic fibrosis model. En-
zyme localization of the ectonucleotidases NTPDase1, -2, -8
and ecto-5=-nucleotidase was determined by immunohisto-
chemistry in liver sections from vehicle (control) and CCl4-
treated rats. In control animals, the expression patterns of
NTPDase1, -2, -8 and ecto-5=-nucleotidase are similar to those
described in Fig. 5. In contrast, in fibrotic animals, NTPDase1
expression is mainly associated with proliferating vascular
structures. NTPDase2 expression is almost exclusively de-
tected in perinodular fibrous areas. NTPDase8 protein is only
found in bile canaliculi, although with a diffuse distribution.
Immunoreactivity to ecto-5=-nucleotidase is localized at the
level of both canalicular and basolateral membrane domains in
hepatocytes, and prominently in fibrous septa surrounding
hepatic fibrotic nodules. Scale bar, 10 ␮m.

adenosine generation was limited only by the rate of AMP dase. This observation is in agreement with the Km value of
production by NTPDases. Of note, under the latter conditions, NTPDase8 for ADP, which is much higher than for NTPDase1
NTPDase8 behaved somewhat like NTPDase2 since it hydro- (18, 31).
lyzed ATP to ADP, with only minimal production of AMP, What functional importance could such a distribution of
which greatly limited adenosine generation by ecto-5=-nucleoti- NTPDases and ecto-5=-nucleotidase have in the liver? At

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES G457

Downloaded from ajpgi.physiology.org on March 1, 2012


Fig. 9. ATP hydrolysis profile by the different combinations of ecto-5=-nucleotidase with hepatic NTPDases. Hydrolysis products of 500 ␮M (high) and 1 ␮M
(low) ATP were analyzed over time, as indicated in EXPERIMENTAL PROCEDURES. At high substrate concentration, ATP is rapidly and completely converted into
adenosine in the presence of both NTPDase1 and ecto-5=-nucleotidase (CD73), whereas adenosine is hardly produced by the NTPDase2 ⫹ ecto-5=-nucleotidase
combination. ATP hydrolysis in the presence of NTPDase8 ⫹ ecto-5=-nucleotidase combination generated a transient accumulation of ADP and AMP that
resulted in a delayed (compared with the NTPDase1 ⫹ ecto-5=-nucleotidase combination) but substantial adenosine production (when compared with the
NTPDase2 ⫹ ecto-5=-nucleotidase combination). At low substrate level, the incubation of ATP with NTPDase1 ⫹ ecto-5=-nucleotidase leads to its rapid
dephosphorylation to adenosine without an increase in ADP level. In the presence of either NTPDase2 or NTPDase8 together with ecto-5=-nucleotidase, the
accumulation of generated ADP was accompanied by poor AMP production because of limited ADP hydrolysis and thus resulted in a modest adenosine
generation by ecto-5=-nucleotidase. ADO, adenosine.

physiological pH, NTPDase1, -2, and -8 enzymatic activities ical conditions. Moreover, this hypothesis supports the notion
account for the major bulk of hepatic ectonucleotidase activity, that NTPDase1, -2, and -8 likely play nonredundant roles in the
degrading nucleoside tri- and diphosphates into their mono- modulation of purinergic signaling, since ATP hydrolysis and
phosphate counterparts, which are essential for the activity of adenosine generation profiles noticeably change as a function
ecto-5=-nucleotidase, the rate-limiting ectoenzyme for adeno- of the NTPDase species (see Fig. 9). Several liver functions
sine formation (5). Thus the biochemical data presented here could be affected by this specific distribution of NTPDases and
demonstrate that the precise expression pattern of the different ecto-5=-nucleotidase: 1) ion transport by cholangiocytes, an
NTPDases in the liver is of physiological relevance because extracellular nucleotide-dependent mechanism that is essential
they can distinctly modulate P1 and P2 receptor activation. to bile formation (20, 37–38, 57); 2) purine salvaging pathways
Indeed, in a given cellular microenvironment, the coupling of in hepatocytes, as the liver represents the main source of
each hepatic NTPDase species with its ecto-5=-nucleotidase nucleo(s/t)ides for extrahepatic tissues deficient in de novo
complementary activity would feed extracellular purinergic purine biosynthesis (11); 3) cell proliferation, an important
mediators for different signal transduction pathways, according feature of liver tissue, which involves P2 receptor signaling in
to the identity of the purinoceptors expressed at the cell hepatocytes, sinusoidal endothelial cells, and portal fibroblasts (6,
membrane. In our assay, we used two different ATP concen- 22, 28, 50); 4) cell death, since stimulation of either P1 or P2
trations, namely 1 ␮M, which is within the range of nucleotide receptors in hepatocytes has been shown to induce apoptosis (52,
levels measured in the conditioned media of liver cell lines 56); and finally 5) inflammation, since hepatocytes, liver immune
(33), and reported for circulating nucleo(s/t)ides (50 nM to 1 cells such as Kupffer cells and T lymphocytes, portal fibroblasts,
␮M) in both sinusoidal blood and bile flows (10), and 500 ␮M as well as hepatic stellate cells express different P1 and P2
ATP, a value close to those potentially reached in the receptors that might modulate their activation and/or deactivation
extracellular medium during inflammatory liver diseases states in liver diseases (4, 13, 17, 41, 54).
(21, 25, 55). Our results show that the substrate concentra- In summary, the specific distribution of NTPDase1, -2, and -8
tion used can influence the net biochemical activity of the in conjunction with ecto-5=-nucleotidase in the liver differentially
different NTPDase/ecto-5=-nucleotidase pairs, suggesting that regulates extracellular nucleo(s/t)ide levels and, therefore, may
the cellular context is an important parameter for the role of also differentially regulate the associated signaling pathways.
these ectonucleotidases in extracellular nucleotide signaling.
Consequently, purinergic receptors expressed in the vicinity of ACKNOWLEDGMENTS
a particular NTPDase/ecto-5=-nucleotidase combination might The authors thank Dr. H. Zimmermann (J. W. Goethe University, Frankfurt,
be differentially regulated depending on the prevailing biolog- Germany) for providing the plasmid encoding for rat ecto-5=-nucleotidase/

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


G458 FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES

CD73 and Dr. M. J. Fernandes and V. Gagné (Rheumatology and Immunology 16. Dranoff JA, Ogawa M, Kruglov EA, Gaca MD, Sévigny J, Robson SC,
Research Centre, CHUL Research Center) for technical assistance with con- Wells RG. Expression of P2Y nucleotide receptors and ectonucleotidases
focal microscopy. We also thank Dr. R. Poulin (Scientific Proofreading and in quiescent and activated rat hepatic stellate cells. Am J Physiol Gastro-
Writing Service, CHUQ Research Center) for editing this paper. intest Liver Physiol 287: G417–G424, 2004.
17. Dranoff JA, Wells RG. Portal fibroblasts: underappreciated mediators of
biliary fibrosis. Hepatology 51: 1438 –1444, 2010.
GRANTS
18. Fausther M, Lecka J, Kukulski F, Lévesque SA, Pelletier J, Zimmer-
This work was supported by the Canadian Institutes of Health Research mann H, Dranoff JA, Sévigny J. Cloning, purification, and identification
(CIHR; MOP-102472 to J. Sévigny) and by the National Institutes of Health of the liver canalicular ecto-ATPase as NTPDase8. Am J Physiol Gastro-
(NIH/NIDDK R01 DK076735, R01 DK070849 and the Yale Liver Center DK intest Liver Physiol 292: G785–G795, 2007.
45710 awards to J. A. Dranoff). M. Fausther was the recipient of a Doctoral 19. Fausther M, Sévigny J. Extracellular nucleosides and nucleotides regu-
Scholarship from the Government of Gabon and is currently receiving the 2011 late liver functions via a complex system of membrane proteins. C R Biol
Roger L. Jenkins Post doctoral Fellowship from the American Liver Founda- 334: 100 –117, 2011.
tion, E. M. Soliman of a Student Research Grant from the Egyptian Govern- 20. Fiorotto R, Spirli C, Fabris L, Cadamuro M, Okolicsanyi L, Strazza-
ment Ministry of Higher Education, G. Kauffenstein of fellowships from the bosco M. Ursodeoxycholic acid stimulates cholangiocyte fluid secretion in
Institut National de la Santé et de la Recherche Médicale in partnership with mice via CFTR-dependent ATP secretion. Gastroenterology 133: 1603–
the Fonds de Recherche en Santé du Québec (FRSQ) and the Heart and Stroke 1613, 2007.
Foundation of Canada in partnership with the CIHR and the Canadian Stroke 21. Franke H, Illes P. Involvement of P2 receptors in the growth and survival
Network, and J. Sévigny of a New Investigator award from the CIHR and a of neurons in the CNS. Pharmacol Ther 109: 297–324, 2006.
Junior 2 Scholarship from the FRSQ. 22. Gonzales E, Julien B, Serriere-Lanneau V, Nicou A, Doignon I,
Lagoudakis L, Garcin I, Azoulay D, Duclos-Vallee JC, Castaing D,
Samuel D, Hernandez-Garcia A, Awad SS, Combettes L, Thevanan-
DISCLOSURES
ther S, Tordjmann T. ATP release after partial hepatectomy regulates

Downloaded from ajpgi.physiology.org on March 1, 2012


No conflicts of interest, financial or otherwise, are declared by the author(s). liver regeneration in the rat. J Hepatol 52: 54 –62, 2010.
23. Hart ML, Gorzolla IC, Schittenhelm J, Robson SC, Eltzschig HK.
SP1-dependent induction of CD39 facilitates hepatic ischemic precondi-
REFERENCES
tioning. J Immunol 184: 4017–4024, 2010.
1. Airas L, Salmi M, Jalkanen S. Lymphocyte-vascular adhesion protein-2 24. Hashimoto M, Kothary PC, Raper SE. Phenobarbital in comparison
is a novel 70-kDa molecule involved in lymphocyte adhesion to vascular with carbon tetrachloride and phenobarbital-induced cirrhosis in rat liver
endothelium. J Immunol 151: 4228 –4238, 1993. regeneration. J Surg Res 81: 164 –169, 1999.
2. Bachmann S, Ramasubbu K. Immunohistochemical colocalization of 25. Hasko G, Linden J, Cronstein B, Pacher P. Adenosine receptors:
the alpha-subunit of neutrophil NADPH oxidase and ecto-5=-nucleotidase therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug
in kidney and liver. Kidney Int 51: 479 –482, 1997. Discov 7: 759 –770, 2008.
3. Baykov AA, Evtushenko OA, Avaeva SM. A malachite green procedure 26. Heine P, Braun N, Heilbronn A, Zimmermann H. Functional charac-
for orthophosphate determination and its use in alkaline phosphatase- terization of rat ecto-ATPase and ecto-ATP diphosphohydrolase after
based enzyme immunoassay. Anal Biochem 171: 266 –270, 1988. heterologous expression in CHO cells. Eur J Biochem 262: 102–107,
4. Beldi G, Banz Y, Kroemer A, Sun X, Wu Y, Graubardt N, Rellstab A, 1999.
Nowak M, Enjyoji K, Li X, Junger WG, Candinas D, Robson SC. 27. Hunsucker SA, Mitchell BS, Spychala J. The 5=-nucleotidases as regu-
Deletion of CD39 on natural killer cells attenuates hepatic ischemia/ lators of nucleotide and drug metabolism. Pharmacol Ther 107: 1–30,
reperfusion injury in mice. Hepatology 51: 1702–1711, 2010. 2005.
5. Beldi G, Enjyoji K, Wu Y, Miller L, Banz Y, Sun X, Robson SC. The 28. Jhandier MN, Kruglov EA, Lavoie EG, Sévigny J, Dranoff JA. Portal
role of purinergic signaling in the liver and in transplantation: effects of fibroblasts regulate the proliferation of bile duct epithelia via expression of
extracellular nucleotides on hepatic graft vascular injury, rejection and NTPDase2. J Biol Chem 280: 22986 –22992, 2005.
metabolism. Front Biosci 13: 2588 –2603, 2008. 29. Kaczmarek E, Koziak K, Sévigny J, Siegel JB, Anrather J, Beaudoin
6. Beldi G, Wu Y, Sun X, Imai M, Enjyoji K, Csizmadia E, Candinas D, AR, Bach FH, Robson SC. Identification and characterization of CD39
Erb L, Robson SC. Regulated catalysis of extracellular nucleotides by vascular ATP diphosphohydrolase. J Biol Chem 271: 33116 –33122, 1996.
vascular CD39/ENTPD1 is required for liver regeneration. Gastroenter- 30. Kegel B, Braun N, Heine P, Maliszewski CR, Zimmermann H. An
ology 135: 1751–1760, 2008. ecto-ATPase and an ecto-ATP diphosphohydrolase are expressed in rat
7. Braun N, Lenz C, Gillardon F, Zimmermann M, Zimmermann H. brain. Neuropharmacology 36: 1189 –1200, 1997.
Focal cerebral ischemia enhances glial expression of ecto-5=-nucleotidase. 31. Kukulski F, Lévesque SA, Lavoie EG, Lecka J, Bigonnesse F, Knowles
Brain Res 766: 213–226, 1997. AF, Robson SC, Kirley TL, Sévigny J. Comparative hydrolysis of P2
8. Braun N, Zhu Y, Krieglstein J, Culmsee C, Zimmermann H. Upregu- receptor agonists by NTPDases 1, 2, 3 and 8. Purinergic Signal 1:
lation of the enzyme chain hydrolyzing extracellular ATP after transient 193–204, 2005.
forebrain ischemia in the rat. J Neurosci 18: 4891–4900, 1998. 32. Lavoie ÉG, Kukulski F, Lévesque SA, Lecka J, Sévigny J. Cloning and
9. Burnstock G. Purine and pyrimidine receptors. Cell Mol Life Sci 64: characterization of mouse nucleoside triphosphate diphosphohydrolase-3.
1471–1483, 2007. Biochem Pharmacol 67: 1917–1926, 2004.
10. Chari RS, Schutz SM, Haebig JE, Shimokura GH, Cotton PB, Fitz 33. Lazarowski ER, Boucher RC, Harden TK. Mechanisms of release of
JG, Meyers WC. Adenosine nucleotides in bile. Am J Physiol Gastroin- nucleotides and integration of their action as P2X- and P2Y-receptor
test Liver Physiol 270: G246 –G252, 1996. activating molecules. Mol Pharmacol 64: 785–795, 2003.
11. Che MX, Gatmaitan Z, Arias IM. Ectonucleotidases, purine nucleoside 34. Lazarowski ER, Tarran R, Grubb BR, van Heusden CA, Okada S,
transporter, and function of the bile canalicular plasma membrane of the Boucher RC. Nucleotide release provides a mechanism for airway surface
hepatocyte. FASEB J 11: 101–108, 1997. liquid homeostasis. J Biol Chem 279: 36855–36864, 2004.
12. Colgan SP, Eltzschig HK, Eckle T, Thompson LF. Physiological roles 35. Lecka J, Rana MS, Sévigny J. Inhibition of vascular ectonucleotidase
for ecto-5=-nucleotidase (CD73). Purinergic Signal 2: 351–360, 2006. activities by the pro-drugs ticlopidine and clopidogrel favours platelet
13. Di Virgilio F, Boeynaems JM, Robson SC. Extracellular nucleotides as aggregation. Br J Pharmacol 161: 1150 –1160, 2010.
negative modulators of immunity. Curr Opin Pharmacol 9: 507–513, 36. Levitt B, Head RJ, Westfall DP. High-pressure liquid chromatographic-
2009. fluorometric detection of adenosine and adenine nucleotides: application
14. Dranoff JA, Kruglov EA, Robson SC, Braun N, Zimmermann H, Sévi- to endogenous content and electrically induced release of adenyl purines
gny J. The ecto-nucleoside triphosphate diphosphohydrolase NTPDase2/ in guinea pig vas deferens. Anal Biochem 137: 93–100, 1984.
CD39L1 is expressed in a novel functional compartment within the liver. 37. Masyuk AI, Gradilone SA, Banales JM, Huang BQ, Masyuk TV, Lee
Hepatology 36: 1135–1144, 2002. SO, Splinter PL, Stroope AJ, Larusso NF. Cholangiocyte primary cilia
15. Dranoff JA, Kruglov EA, Toure J, Braun N, Zimmermann H, Jain D, are chemosensory organelles that detect biliary nucleotides via P2Y12
Knowles AF, Sévigny J. Ectonucleotidase NTPDase2 is selectively purinergic receptors. Am J Physiol Gastrointest Liver Physiol 295: G725–
down-regulated in biliary cirrhosis. J Investig Med 52: 475–482, 2004. G734, 2008.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org


FINE MODULATION OF ADENOSINE FORMATION BY LIVER ECTOENZYMES G459
38. Minagawa N, Nagata J, Shibao K, Masyuk AI, Gomes DA, Rodrigues activity in unfixed cryostat sections of rat liver using a combined light- and
MA, Lesage G, Akiba Y, Kaunitz JD, Ehrlich BE, Larusso NF, electron-microscope procedure. Histochem J 27: 914 –922, 1995.
Nathanson MH. Cyclic AMP regulates bicarbonate secretion in cholan- 48. Stefan C, Jansen S, Bollen M. Modulation of purinergic signaling by
giocytes through release of ATP into bile. Gastroenterology 133: 1592– NPP-type ectophosphodiesterases. Purinergic Signal 2: 361–370, 2006.
1602, 2007. 49. Strater N. Ecto-5=-nucleotidase: Structure function relationships. Puri-
39. Naito Y, Lowenstein JM. 5=-Nucleotidase from rat heart membranes. nergic Signal 2: 343–350, 2006.
Inhibition by adenine nucleotides and related compounds. Biochem J 226: 50. Thevananther S, Sun H, Li D, Arjunan V, Awad SS, Wyllie S,
645–651, 1985. Zimmerman TL, Goss JA, Karpen SJ. Extracellular ATP activates c-jun
40. Peng Z, Borea PA, Varani K, Wilder T, Yee H, Chiriboga L, Black- N-terminal kinase signaling and cell cycle progression in hepatocytes.
burn MR, Azzena G, Resta G, Cronstein BN. Adenosine signaling Hepatology 39: 393–402, 2004.
contributes to ethanol-induced fatty liver in mice. J Clin Invest 119: 51. Tkacz K, Cioroch M, Skladanowski AC, Makarewicz W. The cytotoxic
582–594, 2009. effect of purine riboside on COS-7 cells. Adv Exp Med Biol 486: 355–359,
41. Peng Z, Fernandez P, Wilder T, Yee H, Chiriboga L, Chan ES, 2000.
Cronstein BN. Ecto-5=-nucleotidase (CD73) -mediated extracellular 52. Wen LT, Knowles AF. Extracellular ATP and adenosine induce cell
adenosine production plays a critical role in hepatic fibrosis. FASEB J 22: apoptosis of human hepatoma Li-7A cells via the A3 adenosine receptor.
Br J Pharmacol 140: 1009 –1018, 2003.
2263–2272, 2008.
53. Wood E, Broekman MJ, Kirley TL, Diani-Moore S, Tickner M,
42. Popov Y, Schuppan D. Targeting liver fibrosis: strategies for develop-
Drosopoulos JH, Islam N, Park JI, Marcus AJ, Rifkind AB. Cell-type
ment and validation of antifibrotic therapies. Hepatology 50: 1294 –1306,
specificity of ectonucleotidase expression and upregulation by 2,3,7,8-
2009.
tetrachlorodibenzo-p-dioxin. Arch Biochem Biophys 407: 49 –62, 2002.
43. Robson SC, Schuppan D. Adenosine: tipping the balance towards hepatic 54. Yang P, Han Z, Chen P, Zhu L, Wang S, Hua Z, Zhang J. A
steatosis and fibrosis. J Hepatol 52: 941–943, 2010. contradictory role of A1 adenosine receptor in carbon tetrachloride- and
44. Robson SC, Sévigny J, Zimmermann H. The E-NTPDase family of bile duct ligation-induced liver fibrosis in mice. J Pharmacol Exp Ther
ectonucleotidases: Structure, function, relationships and pathophysiologi- 332: 747–754, 2010.

Downloaded from ajpgi.physiology.org on March 1, 2012


cal significance. Purinergic Signal 2: 409 –430, 2006. 55. Yegutkin GG. Nucleotide- and nucleoside-converting ectoenzymes: Im-
45. Sévigny J, Robson SC, Waelkens E, Csizmadia E, Smith RN, Lem- portant modulators of purinergic signalling cascade. Biochim Biophys Acta
mens R. Identification and characterization of a novel hepatic canalicular 1783: 673–694, 2008.
ATP diphosphohydrolase. J Biol Chem 275: 5640 –5647, 2000. 56. Zoetewij JP, van de Water B, de Bont HJ, Nagelkerke JF. The role of
46. Sévigny J, Sundberg C, Braun N, Guckelberger O, Csizmadia E, Qawi a purinergic P2z receptor in calcium-dependent cell killing of isolated rat
I, Imai M, Zimmermann H, Robson SC. Differential catalytic properties hepatocytes by extracellular adenosine triphosphate. Hepatology 23: 858 –
and vascular topography of murine nucleoside triphosphate diphosphohy- 865, 1996.
drolase 1 (NTPDase1) and NTPDase2 have implications for thromboregu- 57. Zsembery A, Spirli C, Granato A, LaRusso NF, Okolicsanyi L,
lation. Blood 99: 2801–2809, 2002. Crepaldi G, Strazzabosco M. Purinergic regulation of acid/base transport
47. Song J, Bosch KS, Tigchelaar W, Van Den Munckhof RJ, Schellens in human and rat biliary epithelial cell lines. Hepatology 28: 914 –920,
JP, Van Noorden CJ, Frederiks WM. Demonstration of 5=-nucleotidase 1998.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00165.2011 • www.ajpgi.org

You might also like