You are on page 1of 7

Brief Conclusive Report

Exocytosis of azurophil and arginase


1-containing granules by activated
polymorphonuclear neutrophils is required
to inhibit T lymphocyte proliferation
Rita Rotondo,* Maria Bertolotto,† Gaia Barisione,‡ Simonetta Astigiano,‡
Susanna Mandruzzato,§ Luciano Ottonello,† Franco Dallegri,†
Vincenzo Bronte,ⱍⱍ Silvano Ferrini,‡ and Ottavia Barbieri*,‡,1
Departments of *Experimental Medicine and †Internal Medicine, University of Genova, Italy; ‡Department of Translational
Oncology, National Institute for Cancer Research, Genova, Italy; §Department of Oncological and Surgical Sciences, University
of Padova, Italy; and ⱍⱍUniversity Hospital and Department of Pathology, Verona, Italy
RECEIVED NOVEMBER 12, 2009; REVISED JANUARY 26, 2011; ACCEPTED JANUARY 26, 2011; DOI: 10.1189/jlb.1109737

ABSTRACT thine and urea. By depleting the extracellular milieu of l-argi-


ARG1, expressed by human PMNs, inhibits T cell prolif- nine, ARG down-regulates the expression of the CD3␨ chain in
eration by depleting extracellular L-arginine. Here, we T lymphocytes and induces inhibition of T cell proliferation
report that ARG1, released from gelatinase granules by [1]. In humans, enzyme isoform 1 (ARG1) is stored within the
PMNs, is inactive at physiological pH unless activated intracellular granules of PMNs [2], and its exocytosis can be
by factor(s) stored in azurophil granules. Whereas induced by TNF-␣ or IL-8 [3].
ARG1 exocytosis was induced by TNF-␣ or ionomycin,
On these bases, it was advanced that immune suppression
only the latter mediated the release of both granules,
could result from exocytosis of ARG1-containing granules by
resulting in extracellular ARG enzyme activity at physio-
logical pH. Furthermore, after fractionation of the differ- intact PMNs [4]; however, this hypothesis raises some crucial
ent classes of granules, only the mixture of gelatinase issues.
and azurophil granules resulted in ARG1 activity at First, inhibition of T cell proliferation was investigated only
physiological pH. The use of protease inhibitors indi- in models mimicking PMN massive death typical of purulent
cated the involvement of a PMSF- and leupeptin-sus- inflammatory reaction [5] but not during regulated exocytosis
ceptible serine protease in ARG1 processing and acti- by viable PMNs, which is a finely tuned phenomenon. Thus, a
vation. Finally, the supernatant of viable PMNs under-
clear demonstration of inhibition of T cell proliferation by
going frustrated phagocytosis, which mediates
PMNs through ARG1 exocytosis is still missing.
gelatinase and azurophil granule release, inhibited T
cell proliferation through ARG-dependent mechanisms. Second, it was described that ARG1 is stored in PMN gelati-
In vivo, high ARG1 concentrations and increased ARG nase granules as an inactive molecule [4], whereas the enzyme
enzyme activity, sufficient to inhibit T cell proliferation, released upon PMN cell lysis effectively catabolizes l-arginine
were observed in synovial fluids from RA. These find- [5]. Therefore, it is still unclear whether exocytosed ARG1 can
ings suggest that PMNs, recruited at sites of immune effectively catabolize arginine.
complex deposition, induce ARG1-dependent immune Finally, ARG1 catalytic activity largely depends on pH. In-
suppression through concomitant exocytosis of gelati- deed, the curves of pH dependency for this enzyme show that
nase and azurophil granules. J. Leukoc. Biol. 89:
ARG1 enzymatic activity is maximal in a strong alkaline envi-
721–727; 2011.
ronment (pH 9.5–10.5) and negligible at neutral pH [6, 7]. It
is therefore unclear how the enzyme, exocytosed or released
Introduction upon cell lysis, could be effective under physiological condi-
Effective immune suppression can result from the activity of tions and cause immune suppression.
ARG, the enzyme-catalyzing l-arginine degradation to orni- To elucidate these points, we investigate occurrence and
mechanisms of ARG1-dependent immune suppression follow-
ing regulated exocytosis of granule content by viable human
PMNs. We demonstrate that factor(s) stored in azurophil gran-
Abbreviations: ARG⫽arginase, nor-NOHA⫽N-hydroxy-nor-L-arginine,
RA⫽rheumatoid arthritis
1. Correspondence: Embryogenesis and Carcinogenesis in Animal Models,
The online version of this paper, found at www.jleukbio.org, includes National Institute for Cancer Research, Largo Rosanna Benzi 10, 16132
supplemental information. Genova, Italy. E-mail: ottavia.barbieri@istge.it

0741-5400/11/0089-721 © Society for Leukocyte Biology Volume 89, May 2011 Journal of Leukocyte Biology 721
ules enable ARG1, released from gelatinase granules, to catab- lysates were centrifugated on three-layer Percoll gradient, and granule-con-
olize l-arginine effectively at physiological pH and that activity taining bands were collected. All fractions underwent four cycles of freeze
and thawing. To assess the effectiveness of fractionation, each band was
of serine-protease(s) is involved in ARG1 activation. We also
assayed for ARG, MPO, and lactoferrin.
show that exocytosis of these two classes of granules induces
ARG1-dependent inhibition of T cell proliferation. Finally, we
provide evidence that immune suppression via ARG1 released
MPO and lactoferrin detection
by PMNs might occur in RA joints. MPO and lactoferrin were measured following techniques described previ-
ously [12].
Briefly, MPO activity was determined using 0.167 mg/ml O-dianisidine
(Sigma-Aldrich) and 0.1 mM hydrogen peroxide in 50 mM phosphate buf-
MATERIALS AND METHODS fer (pH 6.0). One unit of enzyme activity was defined as that oxidizing 1
␮mol O-dianisidine/min/25°C.
Cell isolation
Lactoferrin concentration was measured using an ELISA sandwich assay
Blood was collected from healthy volunteers, after informed consent, in based on microtiter plates coated with rabbit anti-human lactoferrin IgG
accordance with the Ethics Committee of the National Institute for Cancer (United States Biochemical, Cleveland, OH, USA) and peroxidase-conju-
Research (Genova, Italy) and Declaration of Helsinki. PMNs were isolated gated rabbit anti-human lactoferrin (United States Biochemicals).
by dextran sedimentation, Ficoll separation, and hypotonic lysis of erythro-
cytes [8]. Nonadherent mononuclear cells (PBLs) were also collected. Cell
viability was measured by the LDHe kit (Aniara, Mason, OH, USA).
Western blot analysis
An aliquot of each subcellular fraction was assayed. A total lysate of PMNs,
Patients obtained by five cycles of freeze and thawing, was used as control. Samples
were then resolved under reducing conditions by 10% SDS-PAGE. After
Synovial fluids were obtained after informed consent from affected knees blotting onto nitrocellulose filters (Hybond C-extra, GE Healthcare, Little
of six patients with RA. The mean duration of disease was 3 ⫾ 1 years. Pa- Chalfont, UK), blots were stained using mouse monoclonal IgG1 anti-hu-
tients—three males and three females, age 51 ⫾ 8 years— had not received man ARG1, clone 1.10, or rabbit polyclonal anti-MPO at a 1:1000 dilution
any intra-articular therapy during the 5 weeks before sample collection. (DakoCytomation, Glostrup, Denmark). After washings, blots were incu-
Synovial fluid was also obtained from six patients with gonoarthrosis. Syno- bated with the appropriate secondary HRP-conjugated antibodies (Dako-
vial fluids were centrifuged at 1000 g for 15 min, and the supernatants Cytomation). Bands were visualized by the ECL system (GE Healthcare). In
were removed aseptically and stored at –20°C in aliquots. Prior of ARG ac- another set of experiments, PMNs were lysed in the presence or absence of
tivity, evaluation samples underwent a further centrifugation at 16,000 g for PMSF 2 mM (Sigma-Aldrich), samples were spun at 13,000 RPM for 10
10 min at 4°C. min, and supernatants were collected and incubated for 30 min at 37°C.
Afterward, a Western blot was carried out using goat polyclonal anti-ARG1
ELISA for ARG1 at a 1:1000 dilution (Santa Cruz Biotechnology, Santa Cruz, CA, USA) or
ARG1 protein was measured in supernatant by using the Arginase I ELISA the above-mentioned anti-MPO antiserum. In this case, a total cell lysate
kit (BioVendor, Brno, Czeck Republic). Absorbance at 450 nm was evalu- obtained from normal human liver was used as control.
ated by a spectrophotometer, and ARG1 concentration was calculated on
the basis of a standard curve. Frustrated phagocytosis
A described previously, technique was followed, with some changes [13]. A
Evaluation of ARG activity 5-mg/ml solution of human IgG (Ig Vena N, Sclavo, Siena, Italy) was incu-
ARG activity was measured by spectrophotometry. To assay cell lysates, 4 ⫻ bated for 2 h in 96-well tissue-culture plates. After washing, 2 ⫻ 106 PMNs
106 PMNs in 200 ␮l buffer (150 mM NaCl, 10 mM Tris/HCl, at the indi- in 200 ␮l l-arginine-free DMEM were added to untreated or IgG-coated
cated pH) underwent seven cycles of freezing and thawing. In some tests, plates. After 30 min at 37°C, supernatant was collected.
PMSF, pepstatin A, aprotinin, leupeptin bestatin, phosphoramidon, or cal-
pain inhibitor (all from Sigma-Aldrich, St. Louis, MO, USA) was added. To T cell proliferation
assay supernatants, in some cases, 4 ⫻ 106 PMNs were resuspended in 200
Supernatants of PMNs that underwent frustrated phagocytosis were col-
␮l l-arginine-free DMEM (Sigma-Aldrich) at the indicated pH, incubated
lected and supplemented with B27 (Gibco Invitrogen, Carlsbad, CA, USA)
for 30 min at 37°C in the presence or absence of TNF-␣ (PeproTech, Lon-
at a 1:50 dilution, 100 ␮M l-arginine, 9 ng/ml MnCl2, and in some in-
don, UK) or ionomycin (Serva, Heidelberg, Germany), and then superna-
stances, nor-NOHA (Inalco, Milan, Italy). Afterward, autologous PBLs were
tant was collected; in other cases, 2 ⫻ 106 PMNs were resuspended in 200
added, incubated for 48 h to starve PBL of intracellular l-arginine, and
␮l l-arginine-free DMEM and underwent frustrated phagocytosis for 30
then seeded at 5 ⫻ 104 cells/well in anti-CD3-coated wells. Coating was
min at 37°C, as described below, and afterward, supernatant was collected.
performed by incubating 96-well tissue-culture plates with 50 ␮l/well 500
To assay synovial fluids, 100 ␮l of each sample was added with 100 ␮l of
␮g/ml UCHT1 mAb for 1 h at 37°C, followed by washing. Five days after,
the above-mentioned buffer. All samples were then supplemented with 250
0.5 ␮Ci 3H-thymidine (GE Healthcare) was added, and incorporation was
mM l-arginine and 100 ␮M MnCl2 and incubated at 37°C for 2 h, and
measured after 24 h.
urea production was measured as described [9]. As preliminary analyses
showed that synovial fluids contained urea, each of them was measured
against the same sample not supplemented with l-arginine and MnCl2,
used as “blank.” In some cases, results were controlled by mass spectrome- RESULTS AND DISCUSSION
try, as described [10]. Results from the two techniques coincided; thus,
only results from spectrophotometry are shown herein. Activation of ARG1 at physiological pH requires
concomitant release of gelatinase and azurophil
Subcellular fractionation granules
We followed a technique described previously, with some changes [11]. Previous data indicated that optimal pH for ARG1 from hu-
NaCl (150 mM) was used as medium without protease inhibitors; all proce- man liver ranged between 9.5 and 10.5 [6, 7], and our prelim-
dures were performed at 4°C or in ice bath. After nitrogen cavitation, cell inary titration experiments on ARG activity in PMN lysates

722 Journal of Leukocyte Biology Volume 89, May 2011 www.jleukbio.org


Rotondo et al. PMN exocytosis inhibits T cell proliferation via ARG1

confirmed these findings (Supplemental Fig. 1). As human ing specific granules; and ␤2, containing gelatinase granules.
PMNs do not express ARG2 [3], ARG1 accounts for all of the To verify the achievement of an effective separation of the
ARG activity. We then addressed the relationship between pH three classes of granules, marker enzymes for azurophil
and enzyme activity in lysates and supernatants of PMNs, at (MPO), specific (lactoferrin), and gelatinase (ARG) granules
pH 9.5 and pH 7.5. When lysates of PMNs were assayed for were assessed for each fraction (Fig. 2A). ARG activity was de-
their ARG activity, significant urea production was detected at tected mainly in the ␤2 fraction at pH 9.5 but not at pH 7.5
pH 9.5 and pH 7.5 (Fig. 1A), and the differences in kinetic (Fig. 2B), confirming that the enzyme is stored in gelatinase
curves did not reach statistical significance. Indeed, some granules. To provide further evidence for the localization of
amount of ARG activity was detected at all of the pH values ARG1 in these granules, Western blot analysis was carried out
tested (Supplemental Fig. 1). on the three granule fractions (Fig. 2C). A 41-kDa band, con-
ARG activity was then assayed in supernatants from viable sistent with the ARG1 MW, was detected by anti-ARG1 anti-
PMNs incubated with degranulating stimuli, such as ionomy- body only in the ␤2 fraction. The occurrence of a second
cin, which mobilizes all types of PMN granules [14], or TNF-␣, band of lower MW in granulocyte lysates will be discussed be-
which fails to mobilize azurophil granules [15]. At pH 9.5, a low. MPO protein was present in a large amount only in frac-
dose-dependent increase in enzymatic activity was found with tion ␣, and the low level of MPO detected in the remaining
both stimuli (Fig. 1B), whereas at pH 7.5, ARG activity was de- fractions is most likely a result of contamination by primary
tected only in supernatants of ionomycin-treated PMNs (Fig. granules. When fraction ␤2 was coincubated at pH 7.5 with
1C). However, when ARG1 protein was measured by ELISA, it one of the other fractions, no significant increase in urea pro-
was evident that both stimuli induced similar levels of ARG1 duction was found with fraction ␤1, whereas strong ARG activ-
release in the medium (Fig. 1D). These findings indicate that ity was induced with fraction ␣ (Fig. 2D). These data confirm
ARG1 activity at physiological pH is achieved only in the that ARG1 stored in gelatinase granules is active only at alka-
presence of PMN intracellular factor(s), which are released line pH and that the concomitant release of azurophil gran-
after cell lysis or upon treatment with ionomycin but not ules is required to trigger effective ARG1 activity at physiologi-
TNF-␣. cal pH.
As ionomycin but not TNF-␣ mobilizes azurophil granules, A proteolytic step is likely involved in this process, as specific
we examined further whether these granules contained fac- protease inhibitors reduced ARG activity at pH 7.5 in PMN
tor(s) contributing to ARG1 activity at physiological pH. To lysates (Fig. 3A). The pattern of inhibition indicates the in-
this end, PMNs were lysed, and intracellular granules were sep- volvement of protease(s) resistant to bestatin, calpain inhibi-
arated by centrifugation on a discontinuous Percoll gradient in tor, phosphoramidon, pepstatin, and aprotinin but susceptible
three fractions: ␣, containing azurophil granules; ␤1, contain- to PMSF and leupeptin. These findings make the involvment

A B C
06 PMNs/hour)

1000
800
oles/106 PMNs/hourr)

5000
800
4000 600
600
Urea (nmoles/10

3000
400
400
2000
Urea (nmo

200 200
1000
0
0 0
Untreatted

ate
TNF-α 2 μg//ml
10 μg//ml
50 μg//ml

μM
μM
μM

Lysa
Ionomycin 1 μ
Lysate


20 μ
Untreated

30 60 120
Incubation time (min)

D
Ionomycin Figure 1. Depending on the degranulating stimulus, ARG1 exocytosed by PMNs is ac-
5 μM
tive or inactive at physiological pH. (A) ARG activity at pH 9.5 (e) and pH 7.5 (Œ) was
evaluated in PMN lysates at the indicated times by measuring urea production by spec-
TNF-α trophotometry. Mean ⫾ 1 sd; n ⫽ 6. (B) PMNs were incubated for 30 min with TNF-␣,
10 μg/ml
ionomycin, or medium alone, and supernatants were collected for ARG activity mea-
surement at pH 9.5. For comparison, the amount of ARG activity displayed by lysing
Medium
the same amount of PMNs is shown. Mean ⫾ 1 sd; n ⫽ 3. (C) The above-mentioned
supernatants were tested for ARG activity at pH 7.5. Mean ⫾ 1 sd; n ⫽ 3. (D) ARG1
concentration in the supernatant of TNF-␣- or ionomycin-stimulated PMNs was mea-
0 40 80 120 sured by ELISA. Mean ⫾ 1 sd; n ⫽ 3.
ARG1 concentration (µg/ml)

www.jleukbio.org Volume 89, May 2011 Journal of Leukocyte Biology 723


A 90 B

al enzyme activity
2500

a (nmoles/hour)
80
70 2000
60
Figure 2. Factor(s) stored in azurophil 50 1500
granules are responsible for ARG1 activ- 40
1000
ity at physiological pH. (A) PMN lysate 30

% of tota

Urea
was separated on three-layer Percoll gra- 20 500
dient, and ␣, ␤1, and ␤2 fractions were 10
assayed for ARG, lactoferrin, and MPO. 0 0
ARG and MPO were measured by spec-
trophotometry and lactoferrin by ELISA.
One representative experiment of three ARG Lactoferrin MPO
is shown. (B) ARG activity was measured
on subcellular fractions ␣, ␤1, and ␤2 at C D
pH 7.5 (solid bars) and pH 9.5 (open 41kDa 1750
Arg-1

Urea (nmoles//hour)
bars). Mean ⫾ 1 sd; n ⫽ 3. (C) The pres- 1500
ence of ARG1 and MPO was assessed on 36kDa
1250
␣, ␤1, and ␤2 fractions by Western blot.
1000
Total cell lysate of PMNs was used as con-
750
trol (lane 1). (D) One-hundredth of a ␤2
fraction was incubated with one-hun- 500
dredth of each of the other two fractions, 55kDa 250
MPO
and ARG activity was assayed at pH 7.5. 0
Mean ⫾ 1 sd; n ⫽ 3.
β2

β1
PMNs

Fractions α

of metalloproteases, cystine proteases, and aminopeptidases Supernatant of PMNs activated with nonphagocytable
unlikely and suggest a role for an aprotinin-resistant but immune complexes inhibits T cell proliferation
PMSF- and leupeptin-susceptible serine protease in inducing Simultaneous granule release, also involving azurophil gran-
ARG1 activity at physiological pH. Indeed, the percentage of ules, occurs in vitro when PMNs adhere to a substrate and are
PMSF-induced inhibition of ARG activity in PMN lysates de- then activated or when they attempt to phagocyte large bodies,
creased with the increase in pH (Supplemental Fig. 1). Fur- such as aggregated bacteria or opsonized cells [18, 19], a phe-
thermore, Western blot analysis performed on PMN lysates nomenon called frustrated phagocytosis or reverse endocytosis.
showed two bands, the expected one (41 kDa) and one mi- To mimic this process in vitro, PMNs were allowed to adhere
grating at 36 kDa (Fig. 3B). The latter band could not be de- to tissue-culture wells coated with IgG, and supernatant was
tected when samples were processed in the presence of PMSF, collected. Under this setting, ARG activity was detectable in
suggesting that it was the result of a proteolityc cleavage of supernatants at pH 7.5 (Fig. 4A), demonstrating that ARG1
ARG1 protein by a PMSF-sensitive serine protease. Studies are was not only released but also enabled to catalyze l-arginine at
currently performed in our laboratory to identify this protease physiological pH. Significant amounts of the azurophil granule
and elucidate further ARG1 processing. The finding that the marker MPO were detected in the same supernatants, thus
ARG1 released from gelatinase granules of PMNs is inactive at demonstrating that concurrent exocytosis of this class of gran-
physiological pH, unless activated by factor(s) stored in azuro- ules occurred together with the release of the factor(s) induc-
phil granules, is in agreement with the biological function of ing ARG1 activity at pH 7.5 (Fig. 4B). The presence of small
PMNs. Indeed, the enzymes stored within intracellular gran- amounts of ARG1 enzyme, active at pH 7.5 in the supernatant
ules of PMNs are usually in the form of inactive proenzymes of untreated PMNs (Figs. 1B–D and 4A), can be explained by
that are activated by proteolytic cleavage after being released the spontaneous granule release occurring upon adherence of
[16]. Therefore, the behavior of ARG1 is not surprising, as the PMNs to a plastic substrate during incubation [20]. As cell via-
native form of this enzyme is active at pH values never at- bility, detected by LDH release, was unaffected (data not
tained within our organism. The necessity for ARG1 to be shown), l-arginine catabolism was likely the result of exocytosis
cleaved to exert its function at physiological pH might under- of gelatinase and azurophil granules by living PMNs with no
lie a control mechanism preventing accidental activation of involvment of cell lysis.
the enzyme and consequent immune suppression. As a matter It is conceivable that PMNs exocytosing gelatinase and
of fact, gelatinase granules are easily mobilized, and azurophil azurophil granules might induce immune suppression by the
ones are exocytosed only in a small amount and under partic- released ARG1. Indeed, supernatants of PMNs undergoing
ular conditions [17], which makes extracellular ARG1 activa- frustrated phagocytosis inhibited T cell proliferation (Fig. 4C).
tion a finely tuned phenomenon. To confirm the major role of l-arginine catabolism in this

724 Journal of Leukocyte Biology Volume 89, May 2011 www.jleukbio.org


Rotondo et al. PMN exocytosis inhibits T cell proliferation via ARG1

A PMSF

Leupeptin
A
600

06PMNs/hour)
Phosphoramidon

Calpain inhibitor

Bestatin
400

Urea
Aprotinin

U
(nmoles/10
Pepstatin
200
0 50 100 150
Urea (% of control)

1 2 3 0
B Medium IgG coating
ARG-1 (41 KDa)
B 5
ARG-1 (36 KDa)

U)
MPO (mU
3
+ -

2
Figure 3. Induction of ARG1 activity at physiological pH requires a
proteolytic step. (A) PMNs were lysed in the presence or absence of
100 ␮M pepstatin A, 100 ␮g/ml aprotinin, 40 ␮M bestatin, 20 ␮M cal- 1
pain inhibitor, 10 ␮M phosphoramidon, 50 ␮g/ml leupeptin, or 2 mM
PMSF. After 30 min, ARG activity was tested at pH 7.5 (solid bars) and
pH 9.5 (open bars). Control without protease inhibitors produced 0
728 ⫾ 143 nmoles urea/106 PMN/h. Mean ⫾ 1 sd; n ⫽ 3. (B) PMNs Medium IgG coating
were lysed in the presence (lane 2) or in the absence (lane 3) of 2
mM PMSF, and then Western blot analysis was performed. Total cell
lysate of human liver was used as control (lane 1). C
PBL
phenomenon, T cell inhibition was reduced significantly in the
presence of nor-NOHA, an ARG-specific inhibitor [21]. PBL+ sup resting PMNs
These results provide the first direct demonstration that via-
ble PMNs can inhibit T cell proliferation via the enzymatic PBL + sup PMNs/IgG
activity of exocytosed ARG1.
PBL + sup PMNs/IgG
ARG activity is higher in synovial fluid from RA than + Nor.NOHA
from gonoarthrosis
0 3000 6000 9000 12000
Synchronous release of azurophil and gelatinase granules oc-
curs in PMNs following activation by immune complexes [22, CPM
23]. If the complexes are distributed along a nonphagocytable
surface, exocytosis is observed; when the complexes are phago- Figure 4. Activated PMNs inhibit T cell proliferation via ARG1 activity.
cytosed, some loss of enzymes from the phagocytic vacuoles (A) Ninety-six well tissue-culture plates were incubated with medium
into the extracellular milieu takes place. This release arises or coated with human IgG. PMNs were then added and supernatant
collected after 30 min. ARG activity was measured at pH 7.5. Mean ⫾
from a momentary opening of the vacuole to allow ingestion, 1 sd; n ⫽ 3. (B) The above-mentioned supernatans were assayed for
and it is particularly evident when the phagocytosed particle is MPO activity by spectrophotometry. Mean ⫾ 1 sd; n ⫽ 3. (C) PBLs
relatively large. It is known that the release of proteolytic en- were incubated with supernatant (sup) of PMNs undergoing frustrated
zymes, stored in azurophil granules by PMNs, activated by im- phagocytosis in IgG-coated tissue-culture plates in the presence or ab-
mune complexes, contributes to tissue injury in autoimmune sence of 10 ␮M nor-NOHA, an ARG1 inhibitor. PBLs alone and with
diseases [23]. This is the case of RA, where Igs and immune the supernatant of unstimulated PMNs were tested as control. After
48 h, necessary for depletion of intracellular ARG, PBLs were activated
complexes are tightly bound to the superficial layers of carti-
with immobilized anti-CD3 mAb, and a standard 3H-thymidine incor-
lage [24], thereby providing a solid surface to facilitate PMN poration test was performed. One representative experiment out of
adherence and activation [25]. Furthermore, particulates the three is shown.
formed by Ig aggregates are present in synovial fluid [26].

www.jleukbio.org Volume 89, May 2011 Journal of Leukocyte Biology 725


These conditions pave the way to extracellular release of sec- Besides RA, PMN-associated, ARG1-dependent immune sup-
ondary, gelatinase, and azurophil granules. As the inflamed pression might also play a role in human solid tumors. In this
articular cavity is infiltrated predominantly by PMNs [27], it is context, we have previously found reduced intracellular ARG1
not surprising that the proteolytic enzymes stored in azurophil content in PMNs infiltrating human lung cancer [10], indicat-
granules play a major role in causing tissue-injuring in RA [28, ing a local ARG1 release and immune suppression in the tu-
29]. It is conceivable that together with exocytosis of these en- mor microenvironment, as also demonstrated by others in re-
zymes, release of the factor(s) activating ARG1 might take nal cell carcinoma [32]. If direct evidence of enzyme activity
place. will be provided, ARG1 might become a potential target for
On the basis of these considerations, we measured the en- therapeutic interventions.
zyme concentration and activity in knee synovial fluids from
patients with RA or gonoarthrosis, a noninflammatory joint
disease used as negative control. Even if the difference in AUTHORSHIP
ARG1 concentration between the two groups was not statisti- R.R. performed T cell proliferation and subcellular fraction-
cally significant, four out of six RA synovial fluids contained ation experiments; M.B. performed frustrated phagocytosis
very high concentrations of ARG1 protein (Fig. 5A), whereas and MPO and lactoferrin analysis; G.B. performed ELISA and
synovial fluids from gonoarthrosis patients contained minimal Western blot experiments; S.A. evaluated ARG activity; S.M.
amounts of the enzyme. When the amount of ARG activity at produced mAb anti-ARG1; and L.O., F.D., V.B., S.F., and O.B.
pH 7.5 was measured, the difference between RA and gonoar- designed experiments, provided supervision, analyzed data,
throsis was highly significant (Fig. 5B). Most importantly, in and wrote the paper.
four RA synovial fluids, the amount of ARG activity detected
was well above the levels detected in the supernatant of PMNs
undergoing frustrated phagocytosis (Fig. 4C). ACKNOWLEDGMENTS
This finding indicates that in RA, PMN-dependent exocyto-
sis and activation of ARG1 may occur within the articular cav- This study was supported by research funding from Italian
ity and that ARG1 activity may reach levels capable of inhibit- Ministero della Salute P. Strategico 2006 (V.B. and O.B.) and
ing T cell proliferation. Other cell types probably contribute P. Ordinario 2006 (S.F.); AICR-UK 2008 grant number 08-0518
to inhibition of T cell proliferation in RA joints, as the ARG2 (V.B. and O.B.); Fondazione Carige grant number
isoform is expressed by synoviocytes and macrophages, which 2008.0812.132 (F.D.); and AIRC (S.F.). We thank Dr. Marina
do not exocytose the enzyme [30]. Fabbi for helpful discussion and expert technical assistance.
It has been demonstrated that ARG1, freed upon PMN cell
death, also suppresses NK cell proliferation [31]. ARG1-depen- REFERENCES
dent depletion of l-arginine, resulting from release of azuro- 1. Rodriguez, P. C., Zea, A. H., DeSalvo, J., Culotta, K. S., Zabaleta, J., Qui-
phil and gelatinase granules by activated PMNs, could there- ceno, D. G., Ochoa, J. B., Ochoa, A. C. (2003) L-arginine consumption
by macrophages modulates the expression of CD3 ␨ chain in T lympho-
fore inhibit T and NK cell proliferation. It remains to be eluci- cytes. J. Immunol. 171, 1232–1239.
dated whether in RA patients, this phenomenon might result 2. Munder, M., Mollinedo, F., Calafat, J., Canchado, J., Gil-Lamaignere, C.,
Fuentes, J. M., Luckner, C., Doschko, G., Soler, G., Eichmann, K., Muller,
in down-regulating inflammatory reaction, deranging immune F. M., Ho, A. D., Goerner, M., Modolell, M. (2005) Arginase I is constitu-
response, or inducing both events. tively expressed in human granulocytes and participates in fungicidal ac-
tivity. Blood 105, 2549 –2556.
3. Rotondo, R., Barisione, G., Mastracci, L., Grossi, F., Orengo, A. M., Costa,
A P = 0,0786
B P = 0,0477
R., Truini, M., Fabbi, M., Ferrini, S., Barbieri, O. (2009) IL-8 induces ex-
ocytosis of arginase 1 by neutrophil polymorphonuclears in nonsmall cell
lung cancer. Int. J. Cancer 125, 887– 893.
g/ml)

700 12500
4. Jacobsen, L. C., Theilgaard-Monch, K., Christensen, E. I., Borregaard, N.
ur)
G1 concentration (µg

600
Urea (nmoles/ml/hou

(2007) Arginase 1 is expressed in myelocytes/metamyelocytes and local-


10000
500 ized in gelatinase granules of human neutrophils. Blood 109, 3084 –3087.
5. Munder, M., Schneider, H., Luckner, C., Giese, T., Langhans, C. D., Fu-
400 7500
entes, J. M., Kropf, P., Mueller, I., Kolb, A., Modolell, M., Ho, A. D.
300 (2006) Suppression of T-cell functions by human granulocyte arginase.
5000
Blood 108, 1627–1634.
200
2500
6. Folley, S. J., Greenbaum, A. L. (1948) Determination of the arginase ac-
100 tivities of homogenates of liver and mammary gland: effects of pH and
ARG

0 0
substrate concentration and especially of activation by divalent metal
Gonoarthrosis RA Gonoarthrosis RA ions. Biochem. J. 43, 537–549.
7. Ikemoto, M., Tabata, M., Miyake, T., Kono, T., Mori, M., Totani, M., Mu-
rachi, T. (1990) Expression of human liver arginase in Escherichia coli. Pu-
rification and properties of the product. Biochem. J. 270, 697–703.
Figure 5. Relevant levels of ARG1 protein and ARG activity can be 8. Böyum, A. (1968) Separation of leukocytes from blood and bone mar-
detected in synovial fluids from RA but not from gonoarthrosis. row. Introduction. Scand. J. Clin. Lab. Invest. Suppl. 97, 7.
(A) ARG1 concentration was measured by ELISA in six synovial fluids 9. Bronte, V., Serafini, P., De Santo, C., Marigo, I., Tosello, V., Mazzoni, A.,
from RA and in two from gonoarthrosis. (B) ARG activity was mea- Segal, D. M., Staib, C., Lowel, M., Sutter, G., Colombo, M. P., Zanovello,
P. (2003) IL-4-induced arginase 1 suppresses alloreactive T cells in tu-
sured at pH 7.5 in the above samples. The horizontal line indicates mor-bearing mice. J. Immunol. 170, 270 –278.
the rate of urea production detected in the supernatant of PMNs acti- 10. Rotondo, R., Mastracci, L., Piazza, T., Barisione, G., Fabbi, M., Cas-
vated by frustrated phagocytosis that inhibited T cell proliferation, as sanello, M., Costa, R., Morandi, B., Astigiano, S., Cesario, A., Sormani,
shown in Fig. 3B. Each symbol indicates the same patient in both pan- M. P., Ferlazzo, G., Grossi, F., Ratto, G. B., Ferrini, S., Frumento, G.
(2008) Arginase 2 is expressed by human lung cancer, but it neither in-
els and is the mean of triplicate assay. Statistical analysis was per- duces immune suppression, nor affects disease progression. Int. J. Cancer
formed by unpaired Student’s t test. 123, 1108 –1116.

726 Journal of Leukocyte Biology Volume 89, May 2011 www.jleukbio.org


Rotondo et al. PMN exocytosis inhibits T cell proliferation via ARG1

11. Kjeldsen, L., Sengelov, H., Lollike, K., Nielsen, M. H., Borregaard, N. ing antitumor responses of T lymphocytes infiltrating human prostate
(1994) Isolation and characterization of gelatinase granules from human cancers. J. Exp. Med. 201, 1257–1268.
neutrophils. Blood 83, 1640 –1649. 22. Weissmann, G., Zurier, R. B., Spieler, P. J., Goldstein, I. M. (1971) Mech-
12. Ottonello, L., Tortolina, G., Amelotti, M., Dallegri, F. (1999) Soluble Fas anisms of lysosomal enzyme release from leukocytes exposed to immune
ligand is chemotactic for human neutrophilic polymorphonuclear leuko- complexes and other particles. J. Exp. Med. 134, 149s–165s.
cytes. J. Immunol. 162, 3601–3606. 23. Henson, P. M. (1971) Interaction of cells with immune complexes: ad-
13. Chatham, W. W., Turkiewicz, A., Blackburn Jr., W. D. (1994) Determi- herence, release of constituents, and tissue injury. J. Exp. Med. 134, 114 –
nants of neutrophil HOCl generation: ligand-dependent responses and 135.
the role of surface adhesion. J. Leukoc. Biol. 56, 654 – 660. 24. Jasin, H. E. (1987) Intra-articular antigen-antibody reactions. Rheum. Dis.
14. Faurschou, M., Sorensen, O. E., Johnsen, A. H., Askaa, J., Borregaard, N. Clin. North Am. 13, 179 –189.
(2002) Defensin-rich granules of human neutrophils: characterization of 25. Firestein, G. S. (2003) Evolving concepts of rheumatoid arthritis. Nature
secretory properties. Biochim. Biophys. Acta 1591, 29 –35. 423, 356 –361.
15. Hanlon, W. A., Stolk, J., Davies, P., Humes, J. L., Mumford, R., Bonney, 26. Hasselbacher, P. (1979) Extracellular aggregates of immunoglobulin in
R. J. (1991) rTNF ␣ facilitates human polymorphonuclear leukocyte ad- synovial fluid from rheumatoid arthritis. J. Rheumatol. 6, 374 –380.
herence to fibrinogen matrices with mobilization of specific and tertiary 27. Zvaifler, N. J. (1971) Immunoreactants in rheumatoid synovial effusions.
but not azurophilic granule markers. J. Leukoc. Biol. 50, 43– 48. J. Exp. Med. 134, 276s–285s.
16. Gullberg, U., Bengtsson, N., Bulow, E., Garwicz, D., Lindmark, A., Ol- 28. Moore, A. R., Appelboam, A., Kawabata, K., Da Silva, J. A., D'Cruz, D.,
sson, I. (1999) Processing and targeting of granule proteins in human Gowland, G., Willoughby, D. A. (1999) Destruction of articular cartilage
neutrophils. J. Immunol. Methods 232, 201–210. by ␣ 2 macroglobulin elastase complexes: role in rheumatoid arthritis.
17. Borregaard, N., Cowland, J. B. (1997) Granules of the human neutro- Ann. Rheum. Dis. 58, 109 –113.
philic polymorphonuclear leukocyte. Blood 89, 3503–3521. 29. Kitsis, E., Weissmann, G. (1991) The role of the neutrophil in rheuma-
toid arthritis. Clin. Orthop. Relat. Res. 63–72.
18. Gardiner, E. E., Mok, S. S., Sriratana, A., Robinson, H. C., Veitch, B. J.,
30. Corraliza, I., Moncada, S. (2002) Increased expression of arginase II in
Lowther, D. A., Handley, C. J. (1994) Polymorphonuclear neutrophils
patients with different forms of arthritis. Implications of the regulation of
release 35S-labeled proteoglycans into cartilage during frustrated phago-
nitric oxide. J. Rheumatol. 29, 2261–2265.
cytosis. Eur. J. Biochem. 221, 871– 879. 31. Oberlies, J., Watzl, C., Giese, T., Luckner, C., Kropf, P., Muller, I., Ho,
19. Taichman, N. S., Tsai, C. C., Baehni, P. C., Stoller, N., McArthur, W. P. A. D., Munder, M. (2009) Regulation of NK cell function by human
(1977) Interaction of inflammatory cells and oral microorganisms. IV. In granulocyte arginase. J. Immunol. 182, 5259 –5267.
vitro release of lysosomal constituents from polymorphonuclear leuko- 32. Rodriguez, P. C., Ernstoff, M. S., Hernandez, C., Atkins, M., Zabaleta, J.,
cytes exposed to supragingival and subgingival bacterial plaque. Infect. Sierra, R., Ochoa, A. C. (2009) Arginase I-producing myeloid-derived sup-
Immun. 16, 1013–1023. pressor cells in renal cell carcinoma are a subpopulation of activated
20. Webster, R. O., Wysolmerski, R. B., Lagunoff, D. (1986) Enhancement of granulocytes. Cancer Res. 69, 1553–1560.
human polymorphonuclear leukocyte adherence to plastic and endothe-
lium by phorbol myristate acetate. Comparison with human C5a. Am. J.
Pathol. 125, 369 –378.
21. Bronte, V., Kasic, T., Gri, G., Gallana, K., Borsellino, G., Marigo, I., Battis- KEY WORDS:
tini, L., Iafrate, M., Prayer-Galetti, T., Pagano, F., Viola, A. (2005) Boost- granulocytes 䡠 T cell proliferation 䡠 arginase 䡠 granules

www.jleukbio.org Volume 89, May 2011 Journal of Leukocyte Biology 727

You might also like