ArticlePDF Available

NAD+ Released during Inflammation Participates in T Cell Homeostasis by Inducing ART2-Mediated Death of Naive T Cells In Vivo

Authors:

Abstract and Figures

Mono ADP-ribosyltransferase 2 (ART2) is an ectoenzyme expressed on mouse T lymphocytes, which catalyze the transfer of ADP-ribose groups from NAD(+) onto several target proteins. In vitro, ADP-ribosylation by ART2 activates the P2X7 ATP receptor and is responsible for NAD(+)-induced T cell death (NICD). Yet, the origin of extracellular NAD(+) and the role of NICD in vivo remain elusive. In a model of acute inflammation induced by polyacrylamide beads, we demonstrate release of NAD(+) into exudates during the early phase of the inflammatory response. This leads to T cell depletion in the draining lymph nodes from wild-type and, more severely, from mice lacking the CD38 NAD(+) glycohydrolase, whereas no effect is observed in ART2-deficient animals. Intravenous injection of NAD(+) used to exacerbate NICD in vivo results in fast and dramatic ART2- and P2X7-dependent depletion of CD4+ and CD8+ T lymphocytes, which can affect up to 80% of peripheral T cells in CD38(-/-) mice. This affects mainly naive T cells as most cells surviving in vivo NAD+ treatment exhibit the phenotype of recently activated/memory cells. Consistently, treatment with NAD(+) abolishes primary Ab response to a T-dependent Ag in NICD-susceptible CD38(-/-) mice but has no effect on the secondary response when given several days after priming. Unexpectedly NAD+ treatment improves the response in their wild-type BALB/c counterparts. We propose that NAD(+) released during early inflammation facilitates the expansion of primed T cells, through ART2-driven death of resting cells, thus contributing to the dynamic regulation of T cell homeostasis.
Content may be subject to copyright.
NAD
Released during Inflammation Participates in T Cell
Homeostasis by Inducing ART2-Mediated Death of Naive T
Cells In Vivo
1
Sahil Adriouch,*
†‡
Sandra Hubert,*
Severine Pechberty,* Friedrich Koch-Nolte,
†‡
Friedrich Haag,
†‡
and Michel Seman
2
*
Mono ADP-ribosyltransferase 2 (ART2) is an ectoenzyme expressed on mouse T lymphocytes, which catalyze the transfer of
ADP-ribose groups from NAD
onto several target proteins. In vitro, ADP-ribosylation by ART2 activates the P2X7 ATP
receptor and is responsible for NAD
-induced T cell death (NICD). Yet, the origin of extracellular NAD
and the role of
NICD in vivo remain elusive. In a model of acute inflammation induced by polyacrylamide beads, we demonstrate release
of NAD
into exudates during the early phase of the inflammatory response. This leads to T cell depletion in the draining
lymph nodes from wild-type and, more severely, from mice lacking the CD38 NAD
glycohydrolase, whereas no effect is
observed in ART2-deficient animals. Intravenous injection of NAD
used to exacerbate NICD in vivo results in fast and
dramatic ART2- and P2X7-dependent depletion of CD4
and CD8
T lymphocytes, which can affect up to 80% of peripheral
T cells in CD38
/
mice. This affects mainly naive T cells as most cells surviving in vivo NAD
treatment exhibit the
phenotype of recently activated/memory cells. Consistently, treatment with NAD
abolishes primary Ab response to a
T-dependent Ag in NICD-susceptible CD38
/
mice but has no effect on the secondary response when given several days
after priming. Unexpectedly NAD
treatment improves the response in their wild-type BALB/c counterparts. We propose
that NAD
released during early inflammation facilitates the expansion of primed T cells, through ART2-driven death of
resting cells, thus contributing to the dynamic regulation of T cell homeostasis. The Journal of Immunology, 2007, 179:
186 –194.
Mammalian mono-ADP-ribosyltransferases (ART)
3
constitute a family of ectoenzymes structurally related
to bacterial toxins catalyzing the transfer of the ADP-
ribose group from NAD
onto amino acid residues of target pro-
teins (1–3). ART1–ART4 paralogs are GPI-anchored in the outer
leaflet of the plasma membrane and are thus responsible for unique
posttranslational modifications of proteins in the extracellular
compartment. Like phosphorylation mediated by protein kinases,
ART-mediated modification of proteins by ADP-ribosylation
leads to structural changes that can either inhibit or stimulate
target protein functions. ART activities in vitro can be split into
those relevant to trans-ADP-ribosylation of free soluble targets
such as cytokines and growth factors or proteins produced by
neighboring cells and those relevant to cis-ADP-ribosylation of
proteins present on the surface of the ART-expressing cells (3).
Although ADP-ribosylation of proteins can easily be shown in
vitro (2, 4 6), its reality and role in vivo are still poorly doc-
umented (7, 8).
In the mouse, two of the six ART paralogs, namely, ART2.1
and ART2.2, are expressed on the surface of most mature pe-
ripheral T lymphocytes (1, 9). It was first shown that a GPI-
anchored ART is expressed on the surface of mouse cytotoxic
T cell lines and that incubation of cells with NAD
leads to the
inhibition of both T cell proliferation and cytotoxic activity (6).
This inhibition was associated with ADP-ribosylation of several
membrane proteins such as LFA-1, CD8, CD27, CD43, CD44,
or CD45 (10 –12). Recently, the association of ART2 with lipid
rafts has been shown to focus ART2 on specific targets (13). It
was postulated that ADP-ribosylation of coreceptors inhibits
TCR-signaling by altering receptor association as well as cell
contacts and T cell trafficking (10, 12). We subsequently
showed that incubation with NAD
leads to the rapid induction
of ART-dependent T cell death in vitro detectable at NAD con-
centrations as low as 1
M, a phenomenon that we proposed to
call NAD
-induced T cell death (NICD) for NAD
-induced
cell death (4, 14). We also discovered that NICD results from
the activation of the P2X7 ATP receptor, which is another tar-
get of murine ART2 (14).
P2X7 belongs to the P2X family of ATP-gated ion channels
expressed on different cell types in the immune system including
T lymphocytes (15, 16). P2X7 activation triggers calcium flux,
shedding of CD62L, phosphatidylserine (PS) exposure, opening of
a large nonselective membrane pore and ultimately cell death by
apoptosis (17–19). Incubation of mouse T lymphocytes with
*University Denis Diderot-Paris 7, EA1556, Paris, France;
Institut National de la
Sante´ et de la Recherche Me´dicale Unite´ 519, Faculty of Medicine and Pharmacy,
Rouen, France;
Institute of Immunology, University Medical Center, Hamburg-Ep-
pendorf, Hamburg, Germany
Received for publication December 20, 2006. Accepted for publication April
14, 2007.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by grants from the Ligue Nationale Contre le Cancer, the
Association pour la Recherche sur le Cancer, and the Ministe`re de la Recherche (to
M.S.); the Fondation pour la Recherche Me´dicale (to S.A.), and the Deutsche
Forschungsgemeinschaft (to F.K.-N. and F.H.).
2
Address correspondence and reprint requests to Prof. Michel Seman, Institut Na-
tional de la Sante´ et de la Recherche Me´dicale, U519, Rouen, F-76000, France. E-mail
address: michel.seman@univ-rouen.fr
3
Abbreviations used in this paper: ART, ADP-ribosyltransferase; PI, propidium
iodide; NADase, NAD
glycohydrolase; ADH, alcohol dehydrogenase; PES,
phenazyne ethosulfate; PS, phosphatidylserine; NICD, NAD
-induced T cell death.
Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00
The Journal of Immunology
www.jimmunol.org
micromolar NAD
concentrations induces all these effects,
whereas P2X7 activation by ATP requires millimolar concentra-
tions (14, 17). NICD depends on ART2 and P2X7 as assessed by
the resistance observed with T cells from ART2-deficient or P2X7-
deficient mice (14, 20, 21). Moreover, C57BL/6 T cells, which
harbor a P451L natural mutation in the C-terminal intracytoplas-
mic domain of P2X7 that severely impairs the response to ATP
(22), are relatively resistant also to NICD in vitro, further support-
ing the conclusion that P2X7 is required for NAD
-induced T cell
apoptosis.
The origin of the ecto-NAD
ART-substrate is still poorly
understood. The intracellular NAD
concentration is in the
range of 1 mM, whereas the plasma concentration is in the
range of 0.1
M, below the Km of ARTs (5, 23, 24). It is
speculated that high amounts of ecto-NAD
can be released
during tissue injury as a consequence of cell lysis. However,
nucleotides such as ATP or NAD
also seem to be released by
nonlytic processes under various physiological conditions in-
cluding hypoxia, inflammation, and mechanical or chemical ac-
tivation (25–27). Connexin 43 hemichannels, for instance, me-
diate transmembrane NAD
fluxes in intact cells (25). Hence,
high local NAD
concentrations may be reached under various
physiological and pathophysiological situations which would
permit ADP-ribosylation of membrane proteins on ART-ex-
pressing neighboring cells. The fate of this NAD
is controlled
by ecto-NADases. As recently shown, the ecto-NADase CD38
expressed on B lymphocytes, competes with ARTs for the
NAD
substrate, controls its availability and therefore the level
of ART2-catalyzed ADP-ribosylation on the surface of mouse T
lymphocytes (28). This regulation is likely to occur in vivo in
secondary lymphoid tissues where T and B lymphocytes are in
close contact.
The aim of the present study was to evaluate NAD
release
during inflammation and to explore its consequence on T cell pop-
ulations in vivo. We used a model of inflammation induced by
polyacrylamide beads to investigate NAD
release in exudates
during the course of an acute inflammatory response. We then
explored the effect of NAD
on the different T cell subsets in
lymph nodes drained by the inflammatory site or after systemic
injection of NAD
. Our results show that high amounts of NAD
are released during acute inflammation. They also demonstrate that
NAD
can induce strong T cell depletion in vivo and that cells
surviving NAD
treatment are enriched in activated/memory T
lymphocytes. We finally show that NAD
treatment in vivo affects
Ab production to T dependent Ags providing evidence that NICD
participates in T cell homeostasis in the course of the immune
response.
Materials and Methods
Mice
BALB/c mice were obtained from the Centre d’Elevage Janvier (Le Genest
Saint Isle, France). ART2-deficient mice were generated by standard ho-
mologous recombination procedures and backcrossed for 12 generations
onto the BALB/c background as described elsewhere (20). CD38-deficient
mice were gifts from Dr. F. Lund (Trudeau Institute, New York, NY) (29).
All mice were maintained under standard conditions in the animal quarter
of the Institute Jacques Monod (University Paris 7, Paris, France). Exper-
iments were performed according to European regulations.
Reagents and Abs
ATP, NAD
, propidium iodide (PI), Con A, and Neurospora crassa
NAD
glycohydrolase (NADase) were obtained from Sigma-Aldrich. PE-
and FITC-conjugated anti-B220, anti-CD3, anti-CD4, anti-CD8, anti-
CD25, anti-CD44, anti-CD69, anti-CD62L, anti-CD11b, anti-Gr1, and An-
nexin V
FITC
were from BD Pharmingen. ART2.2-specific Nika 102 mAb
(9) was purified from hybridoma supernatant by affinity chromatography
on protein G-Sepharose (Pharmacia) and conjugated to FITC (Sigma-
Aldrich). P2X7-specific Hano44 mAb was obtained by genetic immuniza-
tion of rats and fusion of spleen cells with Sp2/0 myeloma cells as de-
scribed previously (9, 30). Abs were purified from hybridoma supernatants
and conjugated to Alexa Fluor 488 (Molecular Probes) according to the
manufacturer’s instructions. SRBC were purchased from Eurobio.
Assay for phosphatidylserine exposure and cell death
Single-cell suspensions from lymph nodes were prepared and processed by
flow cytometry on a FACSCalibur instrument using the CellQuest-Pro pro-
gram (BD Biosciences) as described elsewhere (4, 9). B cells were depleted
by magnetic cell separation using anti-B220 Abs (BD Pharmingen) con-
jugated to Adembeads prepared according to the manufacturer’s instruc-
tions (Ademtech). Purity of T cells was always 95% as verified by FACS
analysis using PE-conjugated anti-B220 and FITC-conjugated anti-CD3.
Following treatment with NAD
or ATP for 30 min at 37°C, cells were
washed in RPMI 1640 medium (Invitrogen Life Technologies) and were
resuspended in annexin V binding buffer (BD Pharmingen). They were stained
for 20 min on ice with FITC-conjugated annexin V (1
g/ml) and propidium
iodide (10
g/ml).
Inflammation
Inflammation was induced as previously described (31). Briefly, animals
were shaved and 800
l of a sterile 67% suspension (53 mg dry weight/ml)
of Biogel P100 (Bio-Rad) in PBS was injected s.c. into the bottom of the
back. At given times, pouches were incised on sacrificed animals. The
Biogel mass was collected under a dissecting microscope with a spatula
and resuspended in 0.5 ml of PBS/20 U/ml heparin. Tubes were weighed
before and after addition of the beads to determine the Biogel mass recov-
ered. After vigorous shaking to extract cells and exudate components from
the gel, beads were allowed to sediment and supernatants were collected.
Cells were then recovered by centrifugation and cell-free exudates were
immediately used for NAD
measurement or frozen and kept at 20°C.
Total cells in exudates were counted on a hemocytometer. Cell subpopu-
lations were differentially evaluated by flow cytometry after staining with
PE-conjugated anti-Gr-1 and FITC-conjugated anti-CD11b.
NAD measurements
NAD
concentrations were determined by a cyclic enzymatic assay as
described previously (23). The first step of the cycle is an enzymatic re-
action catalyzed by the NAD
-dependent alcohol dehydrogenase (ADH).
The next steps are nonenzymatic reactions. NADH
2
produced during the
enzymatic step is oxidized by phenazyne ethosulfate (PES) which, once
reduced, is itself oxidized by thiazolyl blue (MTT). Oxidized NAD
and
PES are used in the next cycle. Absorbance of the reduced form of MTT
is measured at 570 nm in a two-chamber spectrophotometer. A “premix”
solution containing 3.2 ml of sodium-1.5 M (pH 7.8) bicine (N,N-bis(2-
hydroxyethyl)glycine) buffer, 2.4 ml of 10 M ethanol, 2.0 ml of 10 mM
MTT, 1.0 ml of 0.2 M EDTA, 0.4 ml of 100 mg/ml BSA, and 1.0 ml of
H
2
O was prepared. Measurements were performed by mixing in the dark
0.3 ml of premix solution with 0.1 ml of 40 mM PES, 0.1 ml of 1 mg/ml
ADH in sodium-bicine buffer (0.1 M; pH 8.8), and 0.1 ml of a standard
NAD
solution or sample. OD was followed over a period of 3–5 min.
NAD
concentration was deduced from standard curves. Determination of
the initial speed of the cyclic reaction, for each known concentration of
NAD
allowed us draw a standard curve: Vf[C], which then permits
accurate calculation of NAD
concentration in samples. We routinely used
initiation by NAD
for inflammatory exudates (colorless in our model) but
initiation by ADH for plasma. All reagents were purchased from
Sigma-Aldrich.
Immunization and Ab titration
Mice were immunized i.p. with 0.2 ml of a 10% sheep erythrocyte sus-
pension (Eurobio) in PBS (32). Serum samples were collected at appro-
priate times and hemagglutination titers were determined by serial dilutions
of 50
l of serum in PBS with 25
l of a 0.4% SRBC suspension.
Data representation and statistical analysis
Results are expressed as the mean SD. Significance was assessed by
Student’s ttest with pat least inferior to 0.05 indicating statistical
significance.
187The Journal of Immunology
Results
NAD
is released during acute inflammation
To evaluate whether NAD
is released during inflammation and to
determine its concentration in inflammatory exudates, we took ad-
vantage of the experimental model of acute inflammation induced
by s.c. injection of Biogel polyacrylamide beads (31, 33). In this
model, surgical recovery of the beads and their suspension in a
defined volume of PBS gives access to the number of cells and to
the real concentration of NAD
trapped per unit of gel mass re-
covered. This method, which is not traumatic for cells, allows the
determination of free NAD
concentration in inflammatory exu-
dates. Wild-type BALB/c mice were injected with Biogel and
sacrificed at various time points to follow the kinetics of NAD
concentration and cellular infiltration during the course of the in-
flammatory reaction (Fig. 1B). Cells recovered in the gel were
counted and NAD
concentration was determined by an enzy-
matic assay. In this model, maximal cell infiltration is achieved
after 24 h and up to 48 h and primarily correspond to neutrophils
as assessed by Gr-1/CD11b staining (Fig. 1A), which are then
progressively replaced by macrophages (31). Remarkably, free
NAD
was detected in exudates. The concentration peaked at 12 h
after induction of inflammation, a time when the cellular response
was still building up (Fig. 1B). This suggests that NAD
is ac-
tively released from cells as an early response to inflammatory
stimuli as opposed to being passively set free as a result of cell
lysis or neutrophil apoptosis.
The influence of CD38 and ART2 in this process was explored
by performing similar experiments in CD38 and ART2-deficient
BALB/c mice. Twenty-four hours after treatment with Biogel, the
number of neutrophils was lower in exudates from CD38
/
mice
compared with wild type (Fig. 2A). This is consistent with the
established role of CD38 in neutrophil chemotaxis and homing in
response to inflammatory stimuli (34). At peak, NAD
concentra-
tion reached 10
M in exudates from CD38
/
BALB/c mice
but 4
M in their wild-type or ART2
/
counterparts (Fig. 2B).
This indicated that the CD38 NADase participates in the clearance
of NAD
at the inflammatory site. Yet, NAD
release presented
the same kinetics as in wild-type animals with a peak at 12 h
preceding massive neutrophil influx in the pouch (Fig. 2C). NAD
release in exudates had no effect on the basal plasma level, which
remained unchanged throughout the inflammatory reaction (Fig.
2Band data not shown). Higher NAD
concentrations were found
in plasma of untreated (data not shown) and Biogel-treated
CD38
/
mice compared with wild-type animals. This may either
reflect elevated steady-state NAD
levels in these animals or have
resulted from NAD
released by hemolysis occurring during
blood collection and preparation.
Inflammation induces NICD in vivo
In vitro, micromolar concentrations of NAD
are sufficient to in-
duce rapid apoptosis of mouse T cells which can be detected by
annexin V/PI staining (4, 14). We, therefore, next tested whether
NAD
present in inflammatory exudates could induce cell death in
vitro. As shown in Fig. 3A, exudates collected after 12 h from
CD38
/
BALB/c mice induced significant apoptosis of purified
lymph node T cells. The effect was limited due to the dilution of
exudates that was necessary to extract the NAD
from the Biogel,
leading to a working concentration in the range of 1.5
M. The
notion that NAD
in inflammatory exudates caused PS flashing is
supported by the finding that PS flashing was inhibited by pretreat-
ment of exudates with exogenous NADase. Moreover, inflammatory
exudates did not induce any detectable annexin V/PI staining of
ART2-deficient T cells (data not shown).
These results prompted us to evaluate the effect of Biogel-in-
duced inflammation on T cells in lymph nodes from wild-type,
CD38
/
, and ART2
/
BALB/c mice. One day after injection of
Biogel beads s.c., in the bottom of the back, draining (inguinal and
para-aortic) and nondraining (axillary) nodes were separately col-
lected. Cells were counted and analyzed by flow cytometry. A
significant diminution of 20% in the number of CD3
T cells
was observed in the draining nodes of wild-type-treated mice but
not in the nondraining axillary nodes of the same animals (Fig.
3B). A more severe reduction of 50% was observed in the drain-
ing nodes from CD38
/
mice but no effect of inflammation was
observed in ART2
/
mice treated under the same conditions
(Fig. 3B). Together, these results demonstrated that NAD
re-
leased during inflammation can reach the draining nodes and can
FIGURE 1. NAD
release during acute inflammation. Inflammatory
pouches were induced in BALB/c mice with 0.8 ml of a 50% Biogel bead
suspension s.c. Cells and exudates were collected at the times indicated. A,
Phenotype of inflammatory cells collected after 24 h and stained with
CD11b/Gr-1 Abs. B, Kinetics of cell infiltration (E) and NAD
release
() in inflammatory exudates (n6 mice/point). Error bars, SD.
FIGURE 2. Comparison of inflammatory response and NAD
release in wild-type, CD38
/
, and ART2
/
BALB/c mice. Inflammation was induced
as described in Fig. 1. A, Total cell number in inflammatory exudates 24 h after treatment with Biogel were determined in wild-type (WT; n11), CD38
/
(n11), and in ART2
/
mice (n7). B, NAD
concentration in plasma and exudates at 12 h (n6 for each group). C, Kinetics of cell infiltration
and NAD
release in exudates from CD38
/
mice (n6). Error bars, SD.
188 NICD IN VIVO
induce T cell depletion in vivo. Consistent with previous reports
(28), CD38 evidently partially protects T cells from the ART2-
dependent deleterious effect of NAD
.
In CD38
/
mice, T cell depletion in the draining nodes was
accompanied by a relative increase in the fraction of CD62L
low
T cells (Fig. 4). This could reflect the resistance of CD62L
low
T
cells to NAD
-induced death and/or shedding of CD62L by
surviving cells as a consequence of P2X7 activation (14). When
10 mg of NAD
was mixed with Biogel, a more pronounced T
cell depletion and CD62L shedding was observed in draining
nodes but also in nondraining ones (Fig. 4). This confirmed that
NAD
released at local inflamed sites can reach draining lymph
nodes (see Fig. 4, Biogel, middle panels) and even nondraining
lymph nodes through lymphatic and blood circulations provided
that the NAD
amount is high enough (see Fig. 4, Biogel
NAD, right panels).
NAD
injection induces rapid T cell depletion in vivo
To further explore the effect of NAD
on T cell populations in
vivo, BALB/c mice were injected i.v. with the maximal tolerated
dose of either ATP (6 mg) or NAD
(10 mg) in 0.2 ml of PBS.
After 24 h, spleen and lymph nodes were recovered and the size of
the T cell population was evaluated by CD3/B220 staining in flow
cytometry. A significant reduction in the percentage of CD3
cells
was observed in lymphoid organs from NAD
-treated mice,
whereas ATP treatment did not have any significant effects (Fig.
5A). The differential effects of ATP and NAD
were consistent
with the higher concentration of ATP compared with NAD
re-
quired to induce T cell death by P2X7 activation in vitro (14) and
with the short half-life of ATP in body fluids due to widely dis-
tributed ecto-ATPases (35, 36). Reduction of the percentage of
CD3
cells was accompanied by a 40% drop in the absolute T cell
count in secondary lymphoid tissues (Fig. 5B). As expected, T cell
depletion was more dramatic in CD38
/
mice compared with
wild-type BALB/c mice, particularly in lymph nodes (Fig. 5C),
whereas NAD
treatment had no effect on T cells in their
ART2
/
counterparts (Fig. 5D). Evidently, NAD
-induced T
cell depletion in vivo is ART2 dependent, in accord with previous
observations in vitro (14, 20). Results in CD38
/
mice are in
agreement with our previous report demonstrating the antagonistic
effect of the CD38 NADase on ART2-mediated T cell death (28).
In CD38
/
BALB/c mice, T cell depletion was clearly detectable
as early as 3 h after NAD
injection and reached its maximum at
70% after 1 day (Fig. 5E). NAD
treatment had little if any
effects on B cell counts in spleen and lymph nodes. The effect on
T lymphocytes was long-lasting and mice had not fully recovered
normal T cell numbers, 1 wk after NAD
treatment. These obser-
vations strongly suggested that NAD
induces rapid T cell death
in vivo as already described in vitro (4).
Cells resistant to NICD exhibit the phenotype of
activated/memory T cells
The effect of treatment with NAD
in vivo on different T cell
subsets was followed by examining cell phenotypes in lymph
nodes 24 h after the injection of NAD
into CD38
/
BALB/c
mice. The results indicate, first, that CD4
cells appear to be more
sensitive to NICD than their CD8
counterparts (Fig. 6,Aand B).
FIGURE 3. NAD
released in exudates induces T cell apoptosis in
vitro and T cell depletion in vivo. A, Induction of apoptosis in BALB/c T
cells by 12-h inflammatory exudates from CD38
/
BALB/c mice. Puri-
fied T cells were incubated with exudates or NAD
solutions in PBS for 30
min at 37°C and then stained with Annexin V
FITC
/PI. Right panel,5Uof
NADase was added to T cells just before addition of the exudate. No gate
was applied from the scatters plots to analyze living and shrinking/dying
cells. The numbers indicated in the lower right quadrant represent the
percentage of apoptotic cells defined as annexin V
/PI
. Results are rep-
resentative of six individual mice from three independent experiments. B,
T cell number in draining (para-aortic and inguinal) and nondraining (ax-
illary) lymph nodes in control or Biogel-treated wild-type (wt; n12),
CD38
/
(n12), and ART2
/
(n6) BALB/c mice. Lymph nodes
were collected 24 h after induction of inflammation and cells were counted.
T cell numbers were deduced from the percentage of CD3
cells estimated
after CD3/B220 staining in flow cytometry. Vertical bars, SD and pvalues
are from Student’s ttest.
FIGURE 4. Analysis of T cells in draining and nondraining lymph nodes in CD38
/
BALB/c mice 24 h after induction of inflammation. Inflammation
was induced with 0.8 ml of a 50% Biogel bead suspension s.c. or Biogel containing 10 mg of NAD
. T cells from draining (para-aortic and inguinal) and
nondraining (axillary) nodes from CD38
/
BALB/c mice were collected after 24 h and stained with anti-CD3-FITC/anti-B220-PE or anti-CD3-FITC/
anti-CD62L-PE mAbs. Panels are representative of six mice from two independent experiments. Right panels, cells were gated on the CD3
population.
189The Journal of Immunology
Indeed, 78 11% of CD4
cells and 52 21% of CD8
cells
were affected in the lymph nodes of these animals 24 h after the
injection of NAD
(p0.003, n10). This could be accounted
for by a difference in ART2 and/or P2X7 density on the plasma
membrane. In accord with a previous report, we show that CD8
T cells express a higher level of ART2.2 on their surface than
CD4
cells as assessed by staining with ART2.2-specific Nika102
mAb (Ref. 9) and Fig. 6C). Yet, CD8
T cells display a lower
P2X7 receptor density than CD4
cells as assessed by P2X7-spe-
cific Hano44 mAb (Fig. 6C). This suggests that susceptibility to
NICD is more influenced by the density of P2X7 than of ART2 on
the cell surface.
Second, cells surviving NAD
treatment in vivo show striking
phenotypic differences to cells of sham-treated mice. Indeed, in-
creased percentages of CD44
high
and CD69
high
along with a de-
creased percentage of CD62L
high
T lymphocytes were observed
among cells resistant to NICD in vivo (Fig. 6D). These markers
characterize the pool of recently activated/memory T cells. Anal-
ysis of cell numbers in vivo reveals that NAD
treatment has no
direct effect on the size of the CD44
high
T cell population and only
a slight effect on the size of the CD69
high
population (Fig. 6E). Yet,
both populations seem to expand 10 days after treatment, a time at
which the peripheral T cell pool is being replenished. Our results
therefore indicate that NAD
treatment has a strong effect on na-
ive T cells and that activated/memory T lymphocytes are resistant
to NICD in vivo.
Analysis of ART2.2 expression after NAD
treatment, using
Nika102 mAb, reveals that a significant fraction of the CD4
cells
and most of the CD8
cells resistant to NAD
in vivo do not
express ART2.2 on their surface (Fig. 7A). Since activated T cells
shed ART2 from their surface (37), this along with their CD44,
CD62L, and CD69 phenotypes supports the conclusion that endo-
genously activated or memory cells are less sensitive to the dele-
terious effect of NAD
. Moreover, CD4
and CD8
cells resistant
FIGURE 5. Injection of free NAD
induces T cell
depletion in vivo. A, BALB/c mice were injected with
0.2 ml of PBS or 0.2 ml of PBS containing 6 mg of ATP
or 10 mg of NAD
. CD3/B220 staining was performed
on spleen and lymph nodes (13 nodes collected) 24 h
after injection. Profiles are representative of at least 12
mice. B, T cell numbers in spleen and lymph nodes from
BALB/c mice 24 h after treatment with NAD
. The
mean value indicated by the horizontal bars correspond
to 60 910
6
and 41 10 10
6
cells in the spleen
of PBS- vs NAD-treated mice (n9, respectively, and
to 24 510
6
and 15 410
6
cells in the lymph
nodes of PBS- vs NAD-treated mice (n12), respec-
tively. Statistical analyses were performed using Stu-
dent’s ttest. C, CD3/B220 staining in CD38
/
BALB/c 24 h after injection of 10 mg of NAD
. Results
are representative of 12 individual mice. D, CD3/B220
staining in ART2
/
BALB/c 24 h after injection of 10
of mg NAD
. Results are representative of five indi-
vidual mice. E, CD38
/
BALB/c mice were treated
with 10 mg of NAD
i.v., and total cell counts were
determined in spleen and lymph nodes at various times.
T and B cell numbers were deduced from the percent-
ages measured after CD3/B220 staining. Each point rep-
resents the mean of six individual mice.
190 NICD IN VIVO
to NICD in vivo are also poorly stained with Hano44 mAb com-
pared with cells from untreated mice (Fig. 7A), consistent with the
notion that sensitivity to NICD is greater for cells expressing high
levels of P2X7. Correlatively, we show that activation of murine T
cells by Con A (Fig. 7B) or by anti-CD3 Abs (data not shown)
leads to a down modulation of both P2X7 and ART2.2 on the
plasma membrane.
Extracellular NAD
influences the immune response to SRBC
The influence of NAD
on the immune response was tested in
wild-type, ART2
/
, and CD38
/
BALB/c mice after immuni-
zation with SRBC. This Ag was selected for several reasons. First,
SRBC Ab response is T dependent and quickly elicits detectable
primary and secondary serum Ab titers. Second, response to SRBC
does not require the use of adjuvants, which are inflammatory and
delay Ag delivery. In a first series of experiments, mice were
treated with 10 mg of NAD
i.v. at the time of immunization with
210
8
SRBC in saline i.p. Mice were bled after 6 days and
primary agglutination titers were determined. A much lower Ab
titer was observed in CD38
/
mice treated with NAD
vs un-
treated mice, whereas NAD
treatment did not show any detect-
able effect on Ab induction in ART2
/
mice (Fig. 8A). This is
consistent with the dramatic T cell depletion induced by NAD
in
CD38
/
animals and the resistance of T cells from ART2
/
mice to NICD in vivo. Conceivably, elimination of most naive T
cells by NAD
in CD38
/
mice affects the recruitment of Ag-
specific cells.
A second group of mice was treated with NAD
on day 4 after
priming, a time when the response was still building up. They were
then boosted with SRBC on day 14 and the secondary response
was tested on day 20. Agglutination titers were compared with
those in mice treated with NAD
on day 0 and to untreated mice.
Results in Fig. 8Bshow that treatment with NAD
on day 4 after
priming did not block the secondary SRBC response in CD38
/
mice or even slightly increased it in comparison to untreated
CD38
/
mice. Notice that the secondary response in mice treated
with NAD
on day 0 was equivalent to the primary one in un-
treated animals. These results further support the notion that NICD
affects naive T cells, whereas activated T cells escape the delete-
rious effect of NAD
.
Remarkably, different results where obtained in wild-type
BALB/c mice which are less sensitive to NICD than their
CD38
/
counterparts. Indeed, treatment with NAD
in these
mice led to a significant improvement of both primary and sec-
ondary responses compared with controls (Fig. 8). This suggests
that NAD
-induced partial T cell depletion of naive T cells, which
likely corresponds to physiological situations in vivo, may facili-
tate the expansion of Ag-primed T cells. Note that NAD
treat-
ment at day 14, i.e., immediately before the secondary antigenic
challenge, had no effect on the secondary response in wild-type or
in CD38
/
BALB/c mice (Fig. 8B). This result is consistent with
the idea that recently primed T cells responsible for the secondary
response are refractory to NICD.
FIGURE 6. T cell phenotype in CD38
/
BALB/c mice after treatment
with NAD
. CD38
/
BALB/c mice were treated with 10 mg of NAD
i.v. or 0.2 ml of PBS. A, Six peripheral lymph nodes were collected at
different times and total cells were counted. Cell numbers were calculated
after CD3-FITC/B220-PE and CD8-PE/CD4-FITC staining. Each point is
the mean of three individual mice. B, CD4:CD8 ratio among B220-de-
pleted lymph node cells 24 h after treatment with NAD
(representative of
seven mice). C, Comparison of P2X7 and ART2.2 expression on CD4
-
and CD8-purified T cells in untreated mice after staining with P2X7-spe-
cific mAb Hano44 or ART2.2-specific mAb Nika102. The mean fluores-
cence intensity (MFI) values were 33 vs 50 for P2X7 expression on CD8
vs CD4
cells, respectively, and 48 vs 21 for ART2.2 expression on CD8
vs CD4
cells, respectively. The MFI values of the isotype controls were
in the range of 5–7. These results are representative of at least four mice.
D, Phenotypes of CD4
lymph node T cells were determined before or
24 h after NAD
injection. Cells were gated on the CD4
population after
staining with CD4-allophycocyanin/CD62L-FITC or CD4-allophycocya-
nin/CD44-CyChrome/CD69-PE. Panels are representative of six individual
mice from three independent experiments. E, Cell numbers were calculated
as in Afrom the percentages observed after CD4/CD44/CD69 staining.
Each point is the mean of three individual mice.
FIGURE 7. ART2 and P2X7 expression after NAD
treatment in vivo.
A, Lymph node-purified T cells from control or NAD
-treated CD38
/
BALB/c mice were stained with PE-conjugated anti-CD4 or anti-CD8 and
counterstained with FITC-conjugated mAb Nika102 mAb for ART2.2 ex-
pression and Alexa Fluor 488-conjugated mAb Hano44 for P2X7 expres-
sion 24 h after treatment with NAD
or PBS. The MFI values in PBS vs
NAD treatment were, respectively, 50 vs 35 (P2X7 expression on CD4
);
33 vs 18 (P2X7 expression on CD8
); 28 vs 23 (ART2.2 expression on
CD4
); and 46 vs 23 (ART2.2 expression on CD8
). The MFI values of
the isotype controls were in the range of 5–7. Panels are representative of
three individual mice. B, Lymph node-purified T cells from wild-type
BALB/c mice were cultured for 24 h with Con A (2
g/ml) in RPMI
1640/10% FCS and stained for expression of P2X7 and ART2.
191The Journal of Immunology
Discussion
The discovery of mono-ART in vertebrates has opened a new field
in cell communication by introducing new ectoenzymes catalyzing
posttranslational modifications of proteins using NAD
in the ex-
tracellular compartment (3). ART1 and ART2 are presently the
two best-characterized paralogs (1), but their role in vivo remains
elusive. In mice, ART2 is expressed on most peripheral T cells and
remarkably induces P2X7 activation by ADP-ribosylation leading
to cell death in vitro (4, 14). Yet, the origin of endogenous extra-
cellular NAD
is still a matter of debate, in which both lytic and
nonlytic processes can be envisioned (25–27). Experiments re-
ported herein provide the first evidence that a significant amount of
NAD
is present in exudates during the early phase of the inflam-
matory response. Inflammation induced by polyacrylamide beads
is Ag free and thus reflects tissue reaction to a foreign bioincom-
patible material. NAD
release precedes massive neutrophil influx
into the inflammatory site. Hence, NAD
release may not be con-
ditioned by massive infiltration of neutrophils. Oxidative stress
induced by activated neutrophils, as well as hypoxia resulting from
the pressure of the Biogel mass under the skin, may contribute to
the liberation of NAD
by neighboring cells through a nonlytic
process. Importantly, the high local NAD
concentration in exu-
dates, which can reach 10
M, has no effect on the plasma
NAD
level. Local NAD
release at an inflammatory site is thus
unlikely to have a global effect on T cell populations in the whole
body. Yet, NAD
concentrations in the range of 3
M were found
in the plasma of CD38
/
mice vs 0.3
M in their wild-type
counterparts. Whether such a high concentration reflects the phys-
iological level in these mice or results from NAD
released in the
absence of the major NAD
-hydrolase CD38 during blood prep-
aration remains questionable. Micromolar NAD
concentrations
are sufficient to induce T cell death in vitro (4) and a NAD
con-
centration in exudates is sufficient to induce detectable NICD in
vitro. Yet, nonmanipulated CD38
/
mice have T cell numbers
and subsets similar to those in wild-type animals. It seems thus
likely that the high NAD
plasma level in CD38
/
animals cor-
responds to an experimental artifact. More importantly, NAD
released at a local inflammatory site induces a significant T cell
depletion in the draining but not in the nondraining nodes from
wild-type or highly sensitive CD38
/
BALB/c mice developing
inflammation to Biogel. This depletion is accompanied by CD62L
shedding on a significant fraction of remaining T cells, an early
event associated with P2X7 activation (18). This, in combination
with T cell resistance in ART2
/
mice, despite NAD
release at
the inflammatory site, provides the first evidence that NICD can
occur under physiopathological situations in vivo.
Consistently, injection of NAD
, the ART2 substrate, into nor-
mal mice expressing both ART2 and P2X7 induces massive T cell
depletion detectable within a few hours following injection. In
support of our findings, accumulation of apoptotic T cells in the
liver has been described in the C57BL/6 mouse strain within 12 h
after NAD
injection (7), although these mice harbor a partially
deficient P2X7 receptor (22). In our experiments, the depletion is
aggravated particularly in CD38
/
mice where it affects 80% of
the T cell population in peripheral lymphoid tissues. No effect of
treatment on the thymus was observed (data not shown), in agree-
ment with the absence of ART2 on thymocytes (38). The high
sensitivity of CD38
/
vs wild-type BALB/c mice to the delete-
rious effect of NAD
is consistent with our previous observation
indicating that the CD38 NADase, mainly expressed on B lym-
phocytes, controls the level of ART2-mediated ADP-ribosylation
of T cell surface proteins and, therefore, T cell apoptosis in un-
fractionated peripheral lymphoid populations (28). The resistance
of T cells from ART2
/
BALB/c mice clearly establishes that T
cell depletion after NAD
treatment results from ART2- and
therefore P2X7-dependent NICD.
The dramatic effect of NAD
injection in vivo not only affects
the number of T lymphocytes in peripheral organs from CD38
/
BALB/c mice but also profoundly influences the distribution of T
cell subpopulations. The fraction of CD44
high
, CD62L
low
and
CD69
high
T cells is strongly enlarged, suggesting that cells resis-
tant to NICD in vivo are activated/memory T cells. Three argu-
ments support this conclusion. First, T cell activation leads to the
shedding of ART2 by the TACE metalloproteinase and renders
cells resistant to NICD (37, 38). Evidently, most of the CD4
and
CD8
T cells present in lymph nodes from NAD
-treated
CD38
/
BALB/c mice do not express ART2.2 on their surface.
Then, most of the CD4
and CD8
cells from NAD
-treated mice
have a low P2X7 density on their surface compared with controls.
We directly show that T cell activation induces a down-modulation
of P2X7 expression and resistance to NICD. This is in agreement
with the low ability of CD44
high
activated/memory CD4
T cells
to open the P2X7 large pore permeable to ethidium bromide in the
presence of ATP (39). Finally, treatment with NAD
at the time of
priming with SRBC inhibits the primary response in CD38
/
BALB/c mice, but treatment on day 4 after priming has no effect
or even stimulates the subsequent response to antigenic challenge
(Fig. 8). This demonstrates that primed T cells escape the delete-
rious effect of NAD
administration. Altogether, these results
demonstrate for the first time that NAD
can modulate the pool of
naive peripheral T cells in vivo, suggesting that endogenous
sources of NAD
may exert a similar effect.
One important aspect of ART2 biology concerns its role in the
regulation of the immune response. Previous experiments have
FIGURE 8. Effect of NAD
treatment on primary and secondary re-
sponses to SRBC. A, To determine the effect of NAD
on the primary
response, CD38
/
, ART2
/
, and wild-type BALB/c mice were primed
with 0.2 ml of a 10% SRBC suspension in PBS i.p. One-half of them
received 10 mg of NAD
in PBS or 0.2 ml of PBS i.v. at the same time as
the SRBC suspension. Agglutination titers were measured on day 6. Each
value represents the mean of six to eight individual mice from two inde-
pendent experiments. B, To determine the effect of NAD
on the secondary
response, mice were primed with SRBC on day 0. For each line, a group
of six mice received 10 mg of NAD
i.v. on day 0, another group on day
4, and the last group received NAD
on day 14. Controls received PBS on
day 0. Mice were boosted with SRBC on day 14 and Ab titers were mea-
sured on day 20. ,p0.05; ⴱⴱ,p0.01 compared to controls.
192 NICD IN VIVO
shown that ART2 ADP-ribosylates several surface proteins includ-
ing LFA-1, CD8, CD27, CD43, CD44, or CD45 (10 –12). This
clearly inhibits cell contact and T cell trafficking (12). ART2-me-
diated ADP-ribosylation of membrane proteins would thus have an
inhibitory effect on the expansion of the T cell response. These
conclusions were drawn from experiments performed in C57BL/6
mice, which express ART2 at a very high density on their T lym-
phocytes (9) but have an impaired P2X7 receptor (22). Conversely,
BALB/c T cells have a lower ART2 density but display a func-
tional P2X7 receptor. Our present results indicate that in the
BALB/c genetic background, CD4
cells, which are ART2
low
P2X7
high
, are more prone to NICD in vivo than CD8
cells which
are ART2
high
P2X7
low
compared to the CD4
.
This suggests that
susceptibility to NICD is governed by P2X7 more than by ART2
density.
Why should NAD
set free in the inflammatory phase of an
infection kill naive T cells during the early development of the
adaptive immune response? This situation, as illustrated by our
experiments in CD38
/
BALB/c mice treated with NAD
at the
time of immunization, could severely impair the response. The
same experiments, however, show that the situation is different in
a normal BALB/c mouse. Priming with NAD
does not impair the
response to Ag but even leads to a weak but significant increase of
the Ab titers. This is also verified during the secondary response
when mice are treated with NAD
4 days after priming. In these
mice, naive T cells are protected from ART2-mediated P2X7
activation and NICD by CD38 expressed on B lymphocytes.
Hence, during the early phase of the immune response, while
activated T cells down-modulate ART2 and P2X7 expression
on their surface, elimination of part of the naive T cells could
give space for the expansion of the Ag-primed T cell population
and thus increase the response. Alternatively, CD4
Foxp3
regulatory T cells may be highly sensitive to NICD, as recently
suggested in C57BL/6 mice (21). Elimination of regulatory T
cells, which exert a negative effect, could contribute toward
improving the response. However, the extent to which the ef-
fects of extracellular NAD
on the development of the immune
response can be attributed to ART-mediated NICD remains
questionable. CD38 is not only a NADase but also an ADP-
ribose cyclase. Cyclic ADP-ribose plays an important role in
the response of neutrophils and professional APCs to chemo-
tactic signals (34, 40). Consequently, CD38
/
mice have a
diminished Ab response to T-dependent Ags (29). The reduced
response to SRBC of CD38
/
vs wild-type BALB/c mice (Fig.
8A) is in fair agreement with this conclusion. Combined with
NICD, a defect in Ag presentation may thus explain the dra-
matic impairment of the primary SRBC response in CD38
/
mice treated with a high dose of NAD
. Similarly, in CD38
/
BALB/c mice, NAD
injection might increase cyclic ADP-ri-
bose production and improve APC activation and trafficking,
compensating the deleterious effect of NICD or even improving
the response. The observation that NAD
administration does
not enhance the primary response in ART2
/
mice, however,
makes it likely that this effect is primarily due to ART2-
mediated NICD.
Taken together, our results demonstrate that NAD
is released
during the early phase of inflammation and illustrate its role in
immune regulation in vivo. They also stress that ART2 and CD38
are two important, but not necessarily opposed partners in this
regulation. Evidently, NICD is a phenomenon which can occur in
vivo. Yet further investigation is needed to define its main target
and its role in the regulation of T cell homeostasis and the control
of autoimmune diseases in normal mice.
Disclosures
The authors have no financial conflict of interest.
References
1. Glowacki, G., R. Braren, K. Firner, M. Nissen, M. Kuhl, P. Reche, F. Bazan,
M. Cetkovic-Cvrlje, E. Leiter, F. Haag, and F. Koch-Nolte. 2002. The family of
toxin-related ecto-ADP-ribosyltransferases in humans and the mouse. Protein
Sci. 11: 1657–1670.
2. Koch-Nolte, F., F. Haag, R. Kastelein, and F. Bazan. 1996. Uncovered: the family
relationship of a T-cell-membrane protein and bacterial toxins. Immunol. Today
17: 402– 405.
3. Seman, M., S. Adriouch, F. Haag, and F. Koch-Nolte. 2004. Ecto-ADP-ribosyl-
transferases (ARTs): emerging actors in cell communication and signaling. Curr.
Med. Chem. 11: 857– 872.
4. Adriouch, S., W. Ohlrogge, F. Haag, F. Koch-Nolte, and M. Seman. 2001. Rapid
induction of naive T cell apoptosis by ecto-nicotinamide adenine dinucleotide:
requirement for mono(ADP-ribosyl)transferase 2 and a downstream effector.
J. Immunol. 167: 196 –203.
5. Krebs, C., W. Koestner, M. Nissen, V. Welge, I. Parusel, F. Malavasi,
E. H. Leiter, R. M. Santella, F. Haag, and F. Koch-Nolte. 2003. Flow cytometric
and immunoblot assays for cell surface ADP-ribosylation using a monoclonal
antibody specific for ethenoadenosine. Anal. Biochem. 314: 108 –115.
6. Wang, J., E. Nemoto, A. Y. Kots, H. R. Kaslow, and G. Dennert. 1994. Regu-
lation of cytotoxic T cells by ecto-nicotinamide adenine dinucleotide (NAD)
correlates with cell surface GPI-anchored/arginine ADP-ribosyltransferase. J. Im-
munol. 153: 4048 4058.
7. Liu, Z. X., O. Azhipa, S. Okamoto, S. Govindarajan, and G. Dennert. 2001.
Extracellular nicotinamide adenine dinucleotide induces t cell apoptosis in vivo
and in vitro. J. Immunol. 167: 4942– 4947.
8. Paone, G., A. Wada, L. A. Stevens, A. Matin, T. Hirayama, R. L. Levine, and
J. Moss. 2002. ADP ribosylation of human neutrophil peptide-1 regulates its
biological properties. Proc. Natl. Acad. Sci. USA 99: 8231– 8235.
9. Koch-Nolte, F., T. Duffy, M. Nissen, S. Kahl, N. Killeen, V. Ablamunits,
F. Haag, and E. H. Leiter. 1999. A new monoclonal antibody detects a develop-
mentally regulated mouse ecto-ADP-ribosyltransferase on T cells: subset distri-
bution, inbred strain variation, and modulation upon T cell activation. J. Immu-
nol. 163: 6014 6022.
10. Liu, Z. X., Y. Yu, and G. Dennert. 1999. A cell surface ADP-ribosyltransferase
modulates T cell receptor association and signaling. J. Biol. Chem. 274:
17399 –17401.
11. Nemoto, E., Y. Yu, and G. Dennert. 1996. Cell surface ADP-ribosyltransferase
regulates lymphocyte function-associated molecule-1 (LFA-1) function in T
cells. J. Immunol. 157: 3341–3349.
12. Okamoto, S., O. Azhipa, Y. Yu, E. Russo, and G. Dennert. 1998. Expression of
ADP-ribosyltransferase on normal T lymphocytes and effects of nicotinamide
adenine dinucleotide on their function. J. Immunol. 160: 4190 4198.
13. Bannas, P., S. Adriouch, S. Kahl, F. Braasch, F. Haag, and F. Koch-Nolte. 2005.
Activity and specificity of toxin-related mouse T cell ecto-ADP-ribosyltrans-
ferase ART2.2 depends on its association with lipid rafts. Blood 105: 3663–3670.
14. Seman, M., S. Adriouch, F. Scheuplein, C. Krebs, D. Freese, G. Glowacki,
P. Deterre, F. Haag, and F. Koch-Nolte. 2003. NAD-induced T cell death: ADP-
ribosylation of cell surface proteins by ART2 activates the cytolytic P2X7 puri-
noceptor. Immunity 19: 571–582.
15. Di Virgilio, F., P. Chiozzi, D. Ferrari, S. Falzoni, J. M. Sanz, A. Morelli,
M. Torboli, G. Bolognesi, and O. R. Baricordi. 2001. Nucleotide receptors: an
emerging family of regulatory molecules in blood cells. Blood 97: 587– 600.
16. North, R. A. 2002. Molecular physiology of P2X receptors. Physiol. Rev. 82:
1013–1067.
17. Surprenant, A., F. Rassendren, E. Kawashima, R. A. North, and G. Buell. 1996.
The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor
(P2X7). Science 272: 735–738.
18. Gu, B., L. J. Bendall, and J. S. Wiley. 1998. Adenosine triphosphate-induced
shedding of CD23 and L-selectin (CD62L) from lymphocytes is mediated by the
same receptor but different metalloproteases. Blood 92: 946 –951.
19. Virginio, C., A. MacKenzie, R. A. North, and A. Surprenant. 1999. Kinetics of
cell lysis, dye uptake and permeability changes in cells expressing the rat P2X7
receptor. J. Physiol. 519: 335–346.
20. Ohlrogge, W., F. Haag, J. Lohler, M. Seman, D. R. Littman, N. Killeen, and
F. Koch-Nolte. 2002. Generation and characterization of ecto-ADP-ribosyltrans-
ferase ART2.1/ART2.2-deficient mice. Mol. Cell. Biol. 22: 7535–7542.
21. Aswad, F., H. Kawamura, and G. Dennert. 2005. High sensitivity of
CD4
CD25
regulatory T cells to extracellular metabolites nicotinamide ade-
nine dinucleotide and ATP: a role for P2X7 receptors. J. Immunol. 175:
3075–3083.
22. Adriouch, S., C. Dox, V. Welge, M. Seman, F. Koch-Nolte, and F. Haag. 2002.
Cutting edge: a natural P451L mutation in the cytoplasmic domain impairs the
function of the mouse P2X7 receptor. J. Immunol. 169: 4108 4112.
23. Jacobson, E. L., and M. K. Jacobson. 1997. Tissue NAD as a biochemical mea-
sure of niacin status in humans. Methods Enzymol. 280: 221–230.
24. Kim, U. H., M. K. Kim, J. S. Kim, M. K. Han, B. H. Park, and H. R. Kim. 1993.
Purification and characterization of NAD glycohydrolase from rabbit erythro-
cytes. Arch. Biochem. Biophys. 305: 147–152.
25. Bruzzone, S., L. Guida, E. Zocchi, L. Franco, and A. De Flora. 2001. Connexin
43 hemi channels mediate Ca
2
-regulated transmembrane NAD
fluxes in intact
cells. FASEB J. 15: 10 –12.
193The Journal of Immunology
26. Di Lisa, F., and M. Ziegler. 2001. Pathophysiological relevance of mitochondria
in NAD
metabolism. FEBS Lett. 492: 4 8.
27. Schwiebert, E. M., and A. Zsembery. 2003. Extracellular ATP as a signaling
molecule for epithelial cells. Biochim. Biophys. Acta 1615: 7–32.
28. Krebs, C., S. Adriouch, F. Braasch, W. Koestner, E. H. Leiter, M. Seman,
F. E. Lund, N. Oppenheimer, F. Haag, and F. Koch-Nolte. 2005. CD38 controls
ADP-ribosyltransferase-2-catalyzed ADP-ribosylation of T cell surface proteins.
J. Immunol. 174: 3298 –3305.
29. Cockayne, D. A., T. Muchamuel, J. C. Grimaldi, H. Muller-Steffner, T. D.
Randall, F. E. Lund, R. Murray, F. Schuber, and M. C. Howard. 1998. Mice
deficient for the ecto-nicotinamide adenine dinucleotide glycohydrolase CD38
exhibit altered humoral immune responses. Blood 92: 1324 –1333.
30. Adriouch, S., G. Dubberke, P. Diessenbacher, F. Rassendren, M. Seman, F. Haag,
and F. Koch-Nolte. 2005. Probing the expression and function of the P2X7 puri-
noceptor with antibodies raised by genetic immunization. Cell. Immunol.
31. Ibanez, O. M., C. Stiffel, O. G. Ribeiro, W. K. Cabrera, S. Massa, M. de Franco,
O. A. Sant’Anna, C. Decreusefond, D. Mouton, M. Siqueira, et al. 1992. Genetics
of nonspecific immunity: I. Bidirectional selective breeding of lines of mice en-
dowed with maximal or minimal inflammatory responsiveness. Eur. J. Immunol.
22: 2555–2563.
32. Seman, M., and V. Zilberfarb. 1979. Genetic control of the IgG2a response to
sheep erythrocytes in mice: isotype- and antigen-specific T cell-mediated sup-
pression in low responders. J. Immunol. 122: 2534 –2540.
33. Ribeiro, O. G., D. A. Maria, S. Adriouch, S. Pechberty, W. H. Cabrera,
J. Morisset, O. M. Ibanez, and M. Seman. 2003. Convergent alteration of gran-
ulopoiesis, chemotactic activity, and neutrophil apoptosis during mouse selection
for high acute inflammatory response. J. Leukocyte Biol. 74: 497–506.
34. Partida-Sanchez, S., D. A. Cockayne, S. Monard, E. L. Jacobson, N.
Oppenheimer, B. Garvy, K. Kusser, S. Goodrich, M. Howard, A. Harmsen, et al.
2001. Cyclic ADP-ribose production by CD38 regulates intracellular calcium
release, extracellular calcium influx and chemotaxis in neutrophils and is required
for bacterial clearance in vivo. Nat. Med. 7: 1209 –1216.
35. Enjyoji, K., J. Sevigny, Y. Lin, P. S. Frenette, P. D. Christie, J. S. Esch II,
M. Imai, J. M. Edelberg, H. Rayburn, M. Lech, et al. 1999. Targeted disruption
of CD39/ATP diphosphohydrolase results in disordered hemostasis and throm-
boregulation. Nat. Med. 5: 1010 –1017.
36. Robson, S. C., Y. Wu, X. Sun, C. Knosalla, K. Dwyer, K. Enjyoji, K. Enjyoji,
J. Sevigny, Y. Lin, P. S. Frenette, et al. 2005. Ectonucleotidases of CD39 family
modulate vascular inflammation and thrombosis in transplantation: targeted dis-
ruption of cd39/ATP diphosphohydrolase results in disordered hemostasis and
thromboregulation. Semin. Thromb. Hemost. 31: 217–233.
37. Kahl, S., M. Nissen, R. Girisch, T. Duffy, E. H. Leiter, F. Haag, and
F. Koch-Nolte. 2000. Metalloprotease-mediated shedding of enzymatically active
mouse ecto-ADP-ribosyltransferase ART2.2 upon T cell activation. J. Immunol.
165: 4463– 4469.
38. Haag, F., D. Freese, F. Scheublein, W. Ohlrogge, S. Adriouch, M. Seman, and
F. Koch-Nolte. 2002. T cells of different developmental stages differ in sensitivity
to apoptosis induced by extracellular NAD. Dev. Immunol. 9: 197–202.
39. Chused, T. M., S. Apasov, and M. Sitkovsky. 1996. Murine T lymphocytes mod-
ulate activity of an ATP-activated P2Z-type purinoceptor during differentiation.
J. Immunol. 157: 1371–1380.
40. Partida-Sanchez, S., T. D. Randall, and F. E. Lund. 2003. Innate immunity is
regulated by CD38, an ecto-enzyme with ADP-ribosyl cyclase activity. Microbes
Infect. 5: 49 –58.
194 NICD IN VIVO
... The selection was based on the observations that a few FAV structural analogues with an amide functionality and an Nheterocyclic ring, including pyrazinamide, temozolomide, and carbamazepine, can cocrystallize with a series of pyridinecarboxamides via formation of amide-amide and amide-pyridine synthons (Sanphui et al., 2013;Aakeroy et al., 2004;Fleischman et al., 2003). In the context of therapeutic potential, NCT and its derivatives function as building blocks for NAD + , which is released during the early stages of inflammation for the purpose of immunomodulation through suppressing the pro-inflammatory cytokines (Adriouch et al., 2007). The neuroprotective (Fricker et al., 2018;Shen et al., 2004) and antiviral (Moëll et al., 2009;Li et al., 2016) effects of NCT are well documented. ...
... One representative experiment out of at least two is shown with n = 3 mice per experiment Abad et al. Journal of Nanobiotechnology (2024) 22:27 potentiating effect was now evident in all analyzed T cell subsets, including in CD8 + T cells that express lower levels of P2X7 [32,33] and are less sensitive than CD4 + subsets ( Fig. 2A, B). Thus, these data show that P2X7 activation in vivo was facilitated by 14D5-dimHLE at low ATP concentrations after i.m. administration of the corresponding AAV vector, AAV-14D5-dimHLE. ...
Article
Full-text available
Background The pro-inflammatory ATP-gated P2X7 receptor is widely expressed by immune and non-immune cells. Nanobodies targeting P2X7, with potentiating or antagonistic effects, have been developed. Adeno-associated virus (AAV)-mediated gene transfer represents an efficient approach to achieve long-term in vivo expression of selected nanobody-based biologics. This approach (AAVnano) was used to validate the relevance of P2X7 as a target in dextran sodium sulfate (DSS)-induced colitis in mice. Results Mice received an intramuscular injection of AAV vectors coding for potentiating (14D5-dimHLE) or antagonistic (13A7-Fc) nanobody-based biologics targeting P2X7. Long-term modulation of P2X7 activity was evaluated ex vivo from blood samples. Colitis was induced with DSS in mice injected with AAV vectors coding for nanobody-based biologics. Severity of colitis, colon histopathology and expression of chemokines and cytokines were determined to evaluate the impact of P2X7 modulation. A single injection of an AAV vector coding for 13A7-Fc or 14D5-dimHLE efficiently modulated P2X7 function in vivo from day 15 up to day 120 post-injection in a dose-dependent manner. An AAV vector coding for 13A7-Fc significantly ameliorated DSS-induced colitis and significantly reduced immune cell infiltration and expression of chemokines and proinflammatory cytokines in colonic tissue. Conclusions We have demonstrated the validity of AAVnano methodology to modulate P2X7 functions in vivo. Applying this methodological approach to a DSS-induced colitis model, we have shown that P2X7 blockade reduces inflammation and disease severity. Hence, this study confirms the importance of P2X7 as a pharmacological target and suggests the use of nanobody-based biologics as potential therapeutics in inflammatory bowel disease. Graphical Abstract
... While CD38 expressed on tumour cells can mediate immune resistance by providing precursors for adenosine generation via the non-canonical pathway mediated by ENPP1 (CD203a) and CD73 (67,68), its NADase function in immune cells is important for protection against ART-mediated NICD under NAD-rich conditions (Figure 1). Indeed, Krebs et al. showed that ART2-mediated mono-ADP-ribosylation following eNAD exposure was elevated in T cells lacking CD38 expression while Adriouch et al. showed that CD38-deficient mice experienced significantly more depletion of P2X7R + T cells following NAD + injection compared to wild-type mice (69,70). ...
Article
Full-text available
While P2X7 receptor expression on tumour cells has been characterized as a promotor of cancer growth and metastasis, its expression by the host immune system is central for orchestration of both innate and adaptive immune responses against cancer. The role of P2X7R in anti-tumour immunity is complex and preclinical studies have described opposing roles of the P2X7R in regulating immune responses against tumours. Therefore, few P2X7R modulators have reached clinical testing in cancer patients. Here, we review the prognostic value of P2X7R in cancer, how P2X7R have been targeted to date in tumour models, and we discuss four aspects of how tumours skew immune responses to promote immune escape via the P2X7R; non-pore functional P2X7Rs, mono-ADP-ribosyltransferases, ectonucleotidases, and immunoregulatory cells. Lastly, we discuss alternative approaches to offset tumour immune escape via P2X7R to enhance immunotherapeutic strategies in cancer patients.
... In human blood, NAD + concentrations were determined to be approximately 33 µM 62 . While this is below the threshold for significant growth inhibition of Spn in our study, concentrations might be higher in the infection-relevant microenvironments like the lung alveoli, especially upon infectioninduced host cell lysis 63 . ...
Article
Full-text available
Lower respiratory tract infections caused by Streptococcus pneumoniae (Spn) are a leading cause of death globally. Here we investigate the bronchial epithelial cellular response to Spn infection on a transcriptomic, proteomic and metabolic level. We found the NAD⁺ salvage pathway to be dysregulated upon infection in a cell line model, primary human lung tissue and in vivo in rodents, leading to a reduced production of NAD⁺. Knockdown of NAD⁺ salvage enzymes (NAMPT, NMNAT1) increased bacterial replication. NAD⁺ treatment of Spn inhibited its growth while growth of other respiratory pathogens improved. Boosting NAD⁺ production increased NAD⁺ levels in immortalized and primary cells and decreased bacterial replication upon infection. NAD⁺ treatment of Spn dysregulated the bacterial metabolism and reduced intrabacterial ATP. Enhancing the bacterial ATP metabolism abolished the antibacterial effect of NAD⁺. Thus, we identified the NAD⁺ salvage pathway as an antibacterial pathway in Spn infections, predicting an antibacterial mechanism of NAD⁺.
... Interestingly, both ATP and NAD + concentrations are low (in the submicromolar range) in the extracellular space, due to the activities of the ectoenzymes CD39 and CD38 that degrade them, respectively [8,35]. Therefore, P2X 7 R activation occurs at inflammatory or damaged sites, as well as in the tumor microenvironment, where ATP and NAD + are released in substantial amounts [36,37]. Accordingly, P2X 7 R-mediated signaling is activated in a large variety of Central Nervous System (CNS) disorders (i.e., Alzheimer's, Parkinson's and Huntington's disease, multiple sclerosis, Amyotrophic Lateral Sclerosis, stroke, neurotrauma, neuropathic pain, epilepsy, and neuropsychiatric disorders), and P2X 7 R antagonists are under intense investigation as a therapy for these conditions (as reviewed in [38]). ...
Article
Full-text available
Extensive evidence indicates that the activation of the P2X7 receptor (P2X7R), an ATP-gated ion channel highly expressed in immune and brain cells, is strictly associated with the release of extracellular vesicles. Through this process, P2X7R-expressing cells regulate non-classical protein secretion and transfer bioactive components to other cells, including misfolded proteins, participating in inflammatory and neurodegenerative diseases. In this review, we summarize and discuss the studies addressing the impact of P2X7R activation on extracellular vesicle release and their activities.
Article
Preeclampsia (PE) is the major cause of maternal-fetal mortality and morbidity. Its pathophysiology is not elucidated, but there is evidence for the role of visfatin/nicotinamide phosphoribosyl transferase (NAMPT), mainly due to its relation to endothelial dysfunction, which is a hallmark of PE. However, there is heterogeneous data regarding visfatin/NAMPT in healthy pregnancy (HP) and PE. Therefore, we performed a search on MEDLINE/PubMed using the terms “visfatin and preeclampsia” and “NAMPT and preeclampsia, and we selected 23 original articles: 12 articles reported increased levels in PE compared to HP, only four articles showed lower levels and eight articles did not find differences regarding visfatin/NAMPT in the groups studied. It is well established that levels detected in plasma, serum, or placenta may suffer the influence of the population and sample size analyzed, along with genetic effects. We further discussed the correlations of visfatin/NAMPT with clinical biomarkers in PE and inflammatory pathways. Considering the common inflammatory mechanisms between PE and visfatin/NAMPT, few studies have recently performed serum or plasma dosages. In conclusion, further studies are needed to highlight the potential role of visfatin/NAMPT in the pathophysiology of PE, which will provide comparative evidence to establish it as a biomarker for disease outcomes and treatment. (Reference: https://doi.org/10.1016/j.jri.2024.104248)
Article
Cancer is a heterogeneous disease. Although both tumor metabolism and tumor immune microenvironment are recognized as driving factors in tumorigenesis, the relationship between them is still not well-known, and potential combined targeting approaches remain to be identified. Here, we demonstrated a negative correlation between the expression of NAMPT, an NAD ⁺ metabolism enzyme, and PD-L1 expression in various cancer cell lines. A clinical study showed that a NAMPT High PD-L1 Low expression pattern predicts poor prognosis in patients with various cancers. In addition, pharmacological inhibition of NAMPT results in the transcription upregulation of PD-L1 by SIRT-mediated acetylation change of NF-κB p65, and blocking PD-L1 would induce NAMPT expression through a HIF-1-dependent glycolysis pathway. Based on these findings, we designed and synthesized a dual NAMPT/PD-L1 targeting compound, LZFPN-90, which inhibits cell growth in a NAMPT-dependent manner and blocks the cell cycle, subsequently inducing apoptosis. Under co-culture conditions, LZFPN-90 treatment contributes to the proliferation and activation of T cells and blocks the growth of cancer cells. Using mice bearing genetically manipulated tumors, we confirmed that LZFPN-90 exerted target-dependent antitumor activities, affecting metabolic processes and the immune system. In conclusion, our results demonstrate the relevance of NAD ⁺ -related metabolic processes in antitumor immunity and suggest that co-targeting NAD ⁺ metabolism and PD-L1 represents a promising therapeutic approach.
Article
Circulating metabolites are indicators of systemic metabolic dysfunction and can be detected through contemporary techniques in metabolomics. These metabolites are involved in numerous mitochondrial metabolic processes including glycolysis, fatty acid β-oxidation, and amino acid catabolism, and changes in the abundance of these metabolites is implicated in the pathogenesis of cardiometabolic diseases (CMDs). Epigenetic regulation and direct metabolite-protein interactions modulate metabolism, both within cells and in the circulation. Dysfunction of multiple mitochondrial components stemming from mitochondrial DNA mutations are implicated in disease pathogenesis. This review will summarize the current state of knowledge regarding: i) the interactions between metabolites found within the mitochondrial environment during CMDs, ii) various metabolites' effects on cellular and systemic function, iii) how harnessing the power of metabolomic analyses represents the next frontier of precision medicine, and iv) how these concepts integrate to expand the clinical potential for translational cardiometabolic medicine.
Article
The extracellular nucleoside adenosine reduces tissue inflammation and is generated by irreversible dephosphorylation of adenosine monophosphate (AMP) mediated by the ectonucleotidase CD73. The pro-inflammatory nucleotides adenosine triphosphate, nicotinamide adenine dinucleotide, and cyclic guanosine -monophosphate-AMP (cGAMP), which are produced in the tumor microenvironment (TME) during therapy-induced immunogenic cell death and activation of innate immune signaling, can be converted into AMP by ectonucleotidases CD39, CD38, and CD203a/ENPP1. Thus, ectonucleotidases shape the TME by converting immune-activating signals into an immunosuppressive one. Ectonucleotidases also hinder the ability of therapies including radiation therapy, which enhance the release of pro-inflammatory nucleotides in the extracellular milieu, to induce immune-mediated tumor rejection. Here, we review the immunosuppressive effects of adenosine and the role of different ectonucleotidases in modulating antitumor immune responses. We discuss emerging opportunities to target adenosine generation and/or its ability to signal via adenosine receptors expressed by immune and cancer cells in the context of combination immunotherapy and radiotherapy.
Article
Full-text available
CD38 is a membrane-associated ecto-nicotinamide adenine dinucleotide (NAD+) glycohydrolase that is expressed on multiple hematopoietic cells. The extracellular domain of CD38 can mediate the catalysis of NAD+ to cyclic adenosine diphosphoribose (cADPR), a Ca2+-mobilizing second messenger, adenosine diphosphoribose (ADPR), and nicotinamide. In addition to its enzymatic properties, murine CD38 has been shown to act as a B-cell coreceptor capable of modulating signals through the B-cell antigen receptor. To investigate the in vivo physiological function(s) of this novel class of ectoenzyme we generated mice carrying a null mutation in the CD38 gene. CD38−/− mice showed a complete loss of tissue-associated NAD+ glycohydrolase activity, showing that the classical NAD+ glycohydrolases and CD38 are likely identical. Although murine CD38 is expressed on hematopoietic stem cells as well as on committed progenitors, we show that CD38 is not required for hematopoiesis or lymphopoiesis. However, CD38−/− mice did exhibit marked deficiencies in antibody responses to T-cell–dependent protein antigens and augmented antibody responses to at least one T-cell–independent type 2 polysaccharide antigen. These data suggest that CD38 may play an important role in vivo in regulating humoral immune responses. © 1998 by The American Society of Hematology.
Article
Full-text available
ADP-ribosyltransferase (ADPRT) is a glycosylphosphatidylinositol-anchored cell surface enzyme on CTL. Expression of this enzyme correlates with suppression of CTL functions in the presence of its substrate β-nicotinamide adenine dinucleotide (NAD). To investigate the immunoregulatory importance of ADPRT on normal lymphocytes in vivo, NAD was injected into mice and the effects on cell-mediated and humoral immunity were assessed. Induction of both delayed-type hypersensitivity and CTL, but not Ab responses, are shown to be suppressed by NAD. Consistent with this, mature T cells, but not B cells or macrophages, express ADPRT and are able to ADP-ribosylate cell surface proteins. ADP-ribosylated molecules were identified as LFA-1, CD8, CD27, CD43, CD44, and CD45. Concomitant to ADP-ribosylation of these molecules, T cell trafficking to secondary lymphoid organs is suppressed by NAD. To examine whether this is due to effects of NAD on cell activation, Ag-stimulated responses were assayed in vitro. NAD is shown to inhibit induction of cell proliferation, cytotoxicity, and cytokine secretion. It is suggested that ADPRT regulates T cells on the level of transmembrane signaling via ADP-ribosylation of cell surface molecules. This effect is reported to be indirect, as it involves transmission of signals through TCRs, which are not ADP-ribosylated.
Article
Full-text available
Cyclic ADP-ribose is believed to be an important calcium-mobilizing second messenger in invertebrate, mammalian and plant cells. CD38, the best-characterized mammalian ADP-ribosyl cyclase, is postulated to be an important source of cyclic ADP-ribose in vivo. Using CD38-deficient mice, we demonstrate that the loss of CD38 renders mice susceptible to bacterial infections due to an inability of CD38-deficient neutrophils to directionally migrate to the site of infection. Furthermore, we show that cyclic ADP-ribose can directly induce intracellular Cal release in neutrophils and is required for sustained extracellular Ca++ influx in neutrophils that have been stimulated by the bacterial chemoattractant, formyl-methionyl-leucyl-phenylalanine (fMLP). Finally, we demonstrate that neutrophil chemotaxis to fMLP is dependent on Cal mobilization mediated by cyclic ADP-ribose. Thus, CD38 controls neutrophil chemotaxis to bacterial chemoattractants through its production of cyclic ADP-ribose, and acts as a critical regulator of inflammation and innate immune responses.
Article
Full-text available
Cyclic ADP-ribose is believed to be an important calcium-mobilizing second messenger in invertebrate, mammalian and plant cells. CD38, the best-characterized mammalian ADP-ribosyl cyclase, is postulated to be an important source of cyclic ADP-ribose in vivo. Using CD38-deficient mice, we demonstrate that the loss of CD38 renders mice susceptible to bacterial infections due to an inability of CD38-deficient neutrophils to directionally migrate to the site of infection. Furthermore, we show that cyclic ADP-ribose can directly induce intracellular Ca++ release in neutrophils and is required for sustained extracellular Ca++ influx in neutrophils that have been stimulated by the bacterial chemoattractant, formyl-methionyl-leucyl-phenylalanine (fMLP). Finally, we demonstrate that neutrophil chemotaxis to fMLP is dependent on Ca++ mobilization mediated by cyclic ADP-ribose. Thus, CD38 controls neutrophil chemotaxis to bacterial chemoattractants through its production of cyclic ADP-ribose, and acts as a critical regulator of inflammation and innate immune responses.
Article
CD23 is a transmembrane protein expressed on the surface of B-lymphocytes that binds IgE, CD21, CD11b, and CD11c. High concentrations of soluble CD23 and L-selectin are found in the serum of patients with B-chronic lymphocytic leukemia (B-CLL). Because extracellular adenosine triphosphate (ATP) causes shedding of L-selectin via activation of P2Z/P2X7 receptors expressed on B-CLL lymphocytes, we studied the effect of ATP on shedding of CD23. ATP-induced shedding of CD23 at an initial rate of 12% of that for L-selectin, whereas the EC50 for ATP was identical (35 μmol/L) for shedding of both molecules. Furthermore, benzoylbenzoyl ATP also produced shedding of CD23 and L-selectin with the same agonist EC50 values for both (10 μmol/L). Inactivation of the P2Z/P2X7 receptor by preincubation with oxidized ATP abolished ATP-induced shedding of both molecules. Moreover, KN-62, the most potent inhibitor for the P2Z/P2X7 receptor, inhibited ATP-induced shedding of both CD23 and L-selectin with the same IC50 (12 nmol/L). Ro 31-9790, a membrane permeant zinc chelator that inhibits the phorbol-ester-stimulated shedding of L-selectin, also inhibited shedding of CD23 from B-CLL lymphocytes. However, the IC50 for this inhibition by Ro31-9790 was different for L-selectin and CD23 (83 v 6 μmol/L, respectively). Although L-selectin was completely shed by incubation of cells with phorbol-ester, CD23 was not lost under these conditions. The data show that extracellular ATP induces shedding of L-selectin and CD23 from B-CLL lymphocytes by an action mediated by the P2Z/P2X7 receptor. However, different membrane metalloproteases seem to mediate the shedding of L-selectin and CD23. © 1998 by The American Society of Hematology.
Article
The known biological roles of niacin are attributable to the function of its active metabolites—nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide phosphate (NADP). In humans, niacin equivalents can be obtained from dietary nicotinate, nicotinamide, and tryptophan. Consequently, niacin status depends on the amount of these in the diet and on factors that influence uptake, distribution, efficiency of conversion to tissue NAD and NADP, and excretion or reutilization of the nicotinamide moiety formed by the turnover of NAD and NADP. Niacin number is chosen as a convenient way to represent niacin status and is defined as NAD/NADP × 100. Expressing the values in this way yields a whole number that is linearly related to intracellular NAD content. Niacin status derived from erythrocytes or whole blood from humans varies over a wide range. The wide range of NAD content is of interest with regard to new questions concerning optimal amounts of dietary niacin raised by the involvement of NAD in ADP-ribose transfer reactions.
Article
ADP-ribosyltransferases including toxins secreted by Vibrio cholera, Pseudomonas aerurginosa, and other pathogenic bacteria inactivate the function of human target proteins by attaching ADP-ribose onto a critical amino acid residue. Cross-species polymerase chain reaction (PCR) and database mining identified the orthologs of these ADP-ribosylating toxins in humans and the mouse. The human genome contains four functional toxin-related ADP-ribosyltransferase genes (ARTs) and two related intron-containing pseudogenes; the mouse has six functional orthologs. The human and mouse ART genes map to chromosomal regions with conserved linkage synteny. The individual ART genes reveal highly restricted expression patterns, which are largely conserved in humans and the mouse. We confirmed the predicted extracellular location of the ART proteins by expressing recombinant ARTs in insect cells. Two human and four mouse ARTs contain the active site motif (R-S-EXE) typical of arginine-specific ADP-ribosyltransferases and exhibit the predicted enzyme activities. Two other human ARTs and their murine orthologues deviate in the active site motif and lack detectable enzyme activity. Conceivably, these ARTs may have acquired a new specificity or function. The position-sensitive iterative database search program PSI-BLAST connected the mammalian ARTs with most known bacterial ADP-ribosylating toxins. In contrast, no related open reading frames occur in the four completed genomes of lower eucaryotes (yeast, worm, fly, and mustard weed). Interestingly, these organisms also lack genes for ADP-ribosylhydrolases, the enzymes that reverse protein ADP-ribosylation. This suggests that the two enzyme families that catalyze reversible mono-ADP-ribosylation either were lost from the genomes of these nonchordata eucaryotes or were subject to horizontal gene transfer between kingdoms.
Article
The IgG2a response to sheep erythrocytes is examined in different congenic strains of mice. B10, B6, and C57BL/Ks animals produce a low level of IgG2a antibodies to SRBC during the primary response in vivo. They remain low responders after secondary challenge in vitro. Total spleen cells or nylon-purified T cells from these low responders inhibit the IgG2a response of H-2 compatible-responding mice in a mixed culture system. This suppression is mediated by Thy-1+, Ly-1-, Ly-2+, and I-J+T cells only present in the spleen of low responding animals. These suppressor T cells appear to be IgG2a- and SRBC-specific. Function of non-H-2-linked genes as regulators of suppressor T cells differentiation is discussed.
Article
The genetic regulation of acute inflammatory reaction (AIR) was studied by the method of bidirectional selective breeding, used to produce a line of mice giving the maximal and a line of mice giving the minimal inflammatory reaction (AIR max and AIR min, respectively). The AIR was triggered by subcutaneous injection of a neutral substrate (suspension of polyacrylamide microbeads), and measured by the leukocyte and serum protein accumulation in the exudate. The two parameters are positively correlated and present a normal frequency distribution. The highly genetically heterogeneous foundation population was produced by the equipoised intercrossing of eight inbred strains of mice, and selective breeding carried out by assortative matings of extreme phenotypes. The response to selection in 11 consecutive generations was highly asymmetrical: a marked AIR increase in the AIR max and no change in the AIR min line occurred. The mean value of realized heritability in the AIR max line was 0.26 and 0.18 for cell and protein concentrations, respectively. The response to selection must have resulted from the interaction of seven to nine independent gene loci endowed with additive effects. The lack of response to selection of the AIR min line is discussed. The large inter-line difference opens new possibilities for studying the biochemistry and molecular genetics of inflammation, and also for investigating the beneficial or detrimental effect of inflammatory responses.
Article
This report demonstrates that incubation of cytotoxic T cells with NAD causes suppression of their ability to proliferate in response to stimulator cells or to lyse targets. Effects are evident after incubation for 3 h with concentrations of NAD as low as 1 microM and are sustained for many hours after removal of NAD from culture media. Suppression is a result of the failure of CTL to form specific conjugates with targets as well as a lower level of activation in response to TCR-mediated stimulation, although TCR-mediated transmembrane signaling is demonstrable. Metabolites of NAD such as nicotinamide, ADP-ribose, and cyclic-ADP-ribose have no detectable effect, indicating that NAD-glycohydrolase or ADP-ribose cyclase do not mediate suppression. Incubation of intact CTL with [32P]NAD leads to incorporation of 32P into a particulate, subcellular fraction, a reaction that is not inhibitable by ADP-ribose. Hydroxylamine, but not mercuric ion releases [32P]ADP-ribose, whereas phosphodiesterase releases [32P]AMP from the particulate subcellular fraction, suggesting that labeling is a result of enzymatic mono-ADP-ribosylation of arginines. In support of this, treatment of intact CTL with phosphatidylinositol-specific phospholipase C releases an arginine-specific ADP-ribosyltransferase and causes insensitivity to ecto-NAD suppression. These results suggest that a GPI-anchored ADP-ribosyltransferase uses ecto-NAD to ADP-ribosylate proteins that regulate CTL function.