You are on page 1of 10

Cell Host & Microbe

Article

Host Inactivation of Bacterial Lipopolysaccharide


Prevents Prolonged Tolerance Following
Gram-Negative Bacterial Infection
Mingfang Lu,1,* Alan W. Varley,1 Shoichiro Ohta,2 John Hardwick,1 and Robert S. Munford1,*
1Infectious Disease Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9113, USA
2Department of Laboratory Medicine, Saga Medical School, 5-1-1 Nabeshima, Saga, Saga 849-8501, Japan

*Correspondence: mingfang.lu@utsouthwestern.edu (M.L.), robert.munford@utsouthwestern.edu (R.S.M.)


DOI 10.1016/j.chom.2008.06.009

SUMMARY Tolerance to LPS has been documented in several clinical set-


tings: adults with pyelonephritis (McCabe, 1963), the alveolar
A transient state of tolerance to microbial molecules macrophages of individuals who have smoked endotoxin-con-
accompanies many infectious diseases. Such toler- taining tobacco (Chen et al., 2007), and the blood leukocytes
ance is thought to minimize inflammation-induced of children with bacterial meningitis (Helminen and Vesikari,
injury, but it may also alter host defenses. Here we 1990a), adults with alcoholic cirrhosis (Von Baehr et al., 2000),
report that recovery from the tolerant state induced and adults with sepsis (McCall et al., 1993; Faist et al., 1988;
Munoz et al., 1991). Perhaps most significantly, tolerance in
by Gram-negative bacteria is greatly delayed in mice
blood leukocytes has been one of the most consistently ob-
that lack acyloxyacyl hydrolase (AOAH), a lipase that
served correlates of infection-induced immunosuppression in
partially deacylates the bacterial cell-wall lipopoly- humans (West and Heagy, 2002). Tolerance has also been noted
saccharide (LPS). Whereas wild-type mice regained in the blood cells of uninfected patients following major trauma
normal responsiveness within 14 days after they re- (West and Heagy, 2002), with greater tolerance noted in those
ceived an intraperitoneal injection of LPS or Gram- who had elevated plasma endotoxin levels (Kelly et al., 1997).
negative bacteria, AOAH-deficient mice had greatly In an investigation of patients with severe sepsis, the blood
reduced proinflammatory responses to a second monocytes of patients with Gram-negative bacterial infections
LPS injection for at least 3 weeks. In contrast, LPS- showed greater tolerance than did those from patients with
primed Aoah- knockout mice maintained an anti-in- Gram-positive infections (Munoz et al., 1991).
flammatory response, evident from their plasma Whereas much is now known about how LPS initiates reprog-
ramming in both monocyte macrophages and neutrophils (Med-
levels of interleukin-10 (IL-10). LPS-primed Aoah-
vedev et al., 2006; Wysocka et al., 2001; Cross, 2002; Cavaillon
knockout mice experiencing prolonged tolerance
et al., 2003; McCall and Yoza, 2007; Foster et al., 2007), the
were highly susceptible to virulent E. coli challenge. mechanisms that terminate the reprogrammed (tolerant) state
Inactivating LPS, an immunostimulatory microbial and restore host defenses are poorly understood (McCall and
molecule, is thus important for restoring effective Yoza, 2007; Wysocka et al., 2005). Indeed, the duration of toler-
host defenses following Gram-negative bacterial ance may vary greatly. In children with Haemophilus influenzae
infection in animals. type b meningitis, leukocyte hyporesponsiveness to LPS re-
solved as the patients recovered over a few days (Helminen
INTRODUCTION and Vesikari, 1990b). In contrast, adult cancer patients, who re-
ceived a bolus intravenous injection of LPS remained tolerant for
As key sensors and effectors of innate immunity, phagocytes almost 2 weeks (Engelhardt et al., 1991), and the peripheral
sense conserved microbial molecules and mount inflammatory blood leukocytes of septic adults have often been reprog-
responses that destroy invading microorganisms. For hours to rammed for many days or weeks after apparently successful
a few days after such an encounter, however, the cells’ proin- treatment of the inciting infection (Weighardt et al., 2000). In sep-
flammatory responses to the same and other microbial mole- tic patients, recovery from tolerance has been associated with
cules may be greatly reduced. This period of tolerance (also survival (Munoz et al., 1991; Weighardt et al., 2000). Understand-
called reprogramming, since some anti-inflammatory responses ing how tolerance is terminated in vivo thus might suggest ways
are usually preserved; Henricson et al. [1993]; Shnyra et al. to decrease the duration of postinfection immunosuppression
[1998]), is thought to minimize inflammation-induced damage and improve outcome.
during recovery from infectious diseases. Although tolerance in- Here we show that recovery from tolerance elicited by LPS or
duced by Gram-negative bacteria or their LPS (endotoxin) has Gram-negative bacteria in vivo requires acyloxyacyl hydrolase
been studied most intensively, a similar phenomenon has also (AOAH), a macrophage, dendritic cell, and neutrophil lipase
been noted in response to other bacterial agonists (Cross, that inactivates LPSs by removing secondary fatty acyl chains
2002; Medvedev et al., 2006; Wang et al., 2003) and during from the lipid A moiety (Munford and Hall, 1986). The duration
some viral infections (Didierlaurent et al., 2008). and degree of tolerance in vivo correlated with the presence of

Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc. 293
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

Figure 1. Aoah / Mice Experience Pro-


longed LPS-Induced Tolerance In Vivo
(A–C) Aoah / and Aoah+/+ mice received intra-
peritoneal injections of PBS or LPS (10 mg) in
PBS on day 0. On days 5, 10, or 22, separate
groups of mice (n = 4–11 mice/group) were chal-
lenged intraperitoneally with 20 mg LPS. Cytokine
or chemokine levels were measured in plasma
samples collected 4 hr later. Combined data
from 4 experiments.
(D and E) Intact Gram-negative bacteria also in-
duce prolonged tolerance in Aoah / mice. Mice
were primed intraperitoneally with 1.5 3 107 col-
ony-forming units of an avirulent strain of E. coli
K12. Fourteen days later they were challenged
intraperitoneally with LPS, and plasma samples
were collected 1 hr (TNF) and 4 hr (IL-6) later for
cytokine analysis (n = 8–10, data combined from
two experiments).
(F) Aoah / mice recover from tolerance induced
by an agonistic antibody to MD-2—TLR4. Mice
were injected intraperitoneally with 1 mg UT12
IgG or heat inactivated (HIA) UT12 on day 0. On
days 5 and 21, separate groups of mice were chal-
lenged intraperitoneally with 20 mg LPS. RANTES
levels were measured in plasma 4 hr after LPS
challenge (n = 4 to 5). Closed symbols, Aoah+/+;
open symbols, Aoah / ; squares, UT12 priming;
triangles, HIA UT12 priming. Similar results were
observed using TNF and IL-6 as response read-
outs (data not shown) and in a second experiment
(n = 4 to 5). Differences from PBS control group
(or HIA UT12 group) of same genotype: * p < 0.05;
**p < 0.01; *** p < 0.001. Error bars represent ± 1
SEM. Note log scales.

fully acylated LPS in peritoneal macrophages. Moreover, mice macrophage reprogramming alters responses along both the
experiencing prolonged tolerance were highly susceptible to MyD88-dependent and -independent arms of the intracellular-
E. coli challenge. These results suggest that killing Gram-nega- signaling pathway downstream of TLR4 (Bagchi et al., 2007;
tive bacteria does not allow full restoration of effective host Sato et al., 2002), in vivo responses to LPS challenge that were
defenses: to prevent prolonged immune suppression, the MyD88-dependent (TNF [Figure S1], IL-6) or MyD88-indepen-
dominant bacterial ‘‘signal’’ molecule, LPS, must also be dent (RANTES from Björkbacka et al., 2004), MCP-1 (Mata-
inactivated. Haro et al., 2007) [Figure S1]) were impaired. AOAH is thus re-
quired for mice to recover their ability to mount several normal
RESULTS proinflammatory responses to LPS. In contrast, LPS-primed
Aoah / mice produced slightly higher plasma concentrations
AOAH Allows Recovery from Tolerance Induced of IL-10, a major anti-inflammatory cytokine, than did the other
by LPS or Gram-Negative Bacteria In Vivo groups (Figure 1C). Retention of a normal IL-10 response to
To investigate a possible role for AOAH in recovery from toler- LPS despite greatly reduced proinflammatory responses sug-
ance, we gave a small intraperitoneal dose of LPS (10 mg) or gests that the tolerant animal’s reactions to LPS are reprog-
PBS to wild-type (Aoah+/+) and AOAH-deficient (Aoah / ) mice rammed, not globally depressed (Henricson et al., 1993; Shnyra
and measured their responses to a second LPS challenge et al., 1998; Medvedev et al., 2006).
(20 mg) given 5, 10, or 22 days later. Aoah / and Aoah+/+ mice Since Gram-negative bacteria also contain nonLPS activators
did not differ in their acute cytokine responses to the first LPS in- of innate immune responses (peptidoglycan, lipoproteins, and
jection (see data for PBS-primed mice in Figures 1A–1C), and the others), it was important to know whether AOAH is also required
mice in both groups were tolerant on day 5 after the initial LPS to terminate tolerance induced by infection with intact Gram-
dose. In contrast, whereas wild-type mice regained their ability negative bacteria. We gave a sublethal infectious dose (1.5 3
to respond normally to a second LPS dose within 5 to 10 days, 107 colony-forming units [cfus]) of an avirulent E. coli strain to
LPS-primed Aoah / mice had greatly reduced plasma IL-6, Aoah+/+ and Aoah / mice and challenged them 14 days later
TNF, and RANTES responses to a second dose for the entire with an intraperitoneal injection of either a virulent E. coli strain
22 day period after the initial LPS exposure (Figures 1A, 1B, (O18K1, 3 3 107 cfus) or 20 mg LPS. As shown in Figures 1D
and S1). In keeping with previous reports that LPS-induced and 1E, Aoah / mice previously exposed to E. coli had

294 Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

significantly lower plasma IL-6 and TNF responses to the LPS


challenge than did Aoah+/+ mice; similar results were seen fol-
lowing virulent E. coli challenge (Figure S2). These findings con-
firm the ability of AOAH to terminate tolerance induced by whole
Gram-negative bacteria and support the conclusion that LPS is
the dominant Gram-negative ‘‘molecular pattern’’ recognized
by animal cells (Somerville et al., 1996; Munford, 2008).

MD-2—TLR4 Activation by an Agonistic Monoclonal


Antibody Does Not Induce Prolonged Tolerance
in AOAH-Deficient Animals
It is possible that AOAH prevents prolonged tolerance through
a mechanism that does not involve its ability to deacylate LPS.
To test this hypothesis, we gave Aoah+/+ and Aoah / mice an
intraperitoneal injection of a monoclonal antibody that stimulates
cells via MD-2—TLR4 in the absence of LPS (Ohta et al., 2006)
and tested their responses to an intraperitoneal dose of LPS
given 5 days or 21 days later. We found identical responses for
the two genotypes: in both Aoah+/+ and Aoah / mice, tolerance
on day 5 was followed by recovery by day 21 (Figure 1F, com-
pare with the responses to a priming dose of LPS in Figure 1B).
AOAH is thus important for recovery from tolerance induced by
its substrate, LPS, but not from tolerance induced by a nonLPS
agonist that activates the same signaling pathway.

LPS-Induced Antibodies Are Not Required to Prolong


Tolerance in AOAH-Deficient Mice
Since Aoah / mice have greatly exaggerated polyclonal IgM
and IgG3 antibody responses to LPS (Lu et al., 2005), we tested
the hypothesis that antibody-mediated LPS neutralization pre-
vents responses to the second LPS challenge in primed animals
(Greisman et al., 1969). Mice that lack both B cells (Kitamura
et al., 1991) and AOAH (mMT, Aoah double knockouts) exhibited
prolonged LPS tolerance in the absence of circulating IgM and
Figure 2. Prolonged Tolerance Is Not Antibody Mediated
IgG (Figure 2). B cells or antibody responses to the priming
(A and B) Aoah / , mMT, and Aoah / mMT mice were primed with LPS intra-
LPS thus do not account for persistent LPS tolerance in peritoneally on day 0 and challenged with LPS on day 14. Plasma samples
Aoah / mice. were collected 4 hr later, and levels of IL-6 (A) and IL-10 (B) were measured
by ELISA. Combined data from three experiments, n = 5–15 mice/group, are
Peritoneal Macrophages Also Exhibit Reprogrammed shown. Error bars represent ± 1 SEM. The differences from PBS control group
Responses to Microbial Ligands of the same genotype are *** p < 0.001.
Macrophages harvested from the peritoneal cavities of LPS-
primed Aoah / mice were also reprogrammed. Re-exposure In Aoah / Animals, Tolerance Can Be Induced by Very
to LPS ex vivo was associated with greatly diminished LPS-in- Low LPS Doses and Last for Two Months
duced MyD88-dependent (TNF, IL-6, IL-1b [data not shown]) LPS-induced tolerance was dose-dependent (Figure 3D); as little
and MyD88-independent (RANTES, nitric oxide production) as 0.1 mg intraperitoneal LPS induced tolerance for 2 weeks in
responses (Figures 3A and 3B). In addition, the production Aoah / peritoneal macrophages, whereas 10 mg or more was
of TNF, IL-6 (data not shown), and RANTES was reduced in required in Aoah+/+ animals. Moreover, tolerance lasted for at
response to Micrococcus luteus, a TLR2- dependent stimulus, least 2 months in the absence of AOAH (Figure 3E). Aoah /
and there was significantly impaired RANTES production after mice of another genetic background (C3H/HeN) also were toler-
stimulation with a MyD88-independent TLR3 agonist, poly I:C ant for 14 days or more following exposure to 10 mg LPS intraper-
(Alexopoulou et al., 2001) (Figure 3A). LPS thus induced pro- itoneally, excluding a strain-specific effect of the Aoah / muta-
longed cross-tolerance (or ‘‘heterotolerance,’’ Dobrovolskaia tion (data not shown).
et al. [2003]) to other microbial agonists in Aoah / animals. In
contrast, whereas LPS-primed Aoah / macrophages produced Recombinant AOAH Prevents Prolonged Tolerance
wild-type levels of IL-10 when incubated with Micrococcus in Aoah / Animals
luteus, their IL-10 response to LPS was approximately one-third To confirm that AOAH deficiency allows prolonged LPS-induced
that of macrophages from primed Aoah+/+ mice (Figure 3C). A tolerance, we expressed recombinant human AOAH in AOAH-
more extensive survey of peritoneal macrophage responses to deficient mice using a gutted adenoviral vector, Ad.CMV-
LPS is presented in Table S1. hAOAH. Fourteen days after viral infection, the mice were given

Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc. 295
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

/
Figure 3. Reprogramming Persists in Explanted Peritoneal Macrophages from LPS-Primed Aoah Mice
(A–C) Aoah / and Aoah+/+ mice were given LPS i.p. (primed mice) or PBS (control mice) on day 0 and on day 10. Ten days after the second injection, peritoneal
macrophages were isolated and treated ex vivo with LPS, Micrococcus luteus (ML), or poly I:C for 6 hr (A and C) or 24 hr (B). RANTES (A), nitrite (B), and IL-10 (C)
were measured in culture supernatants.
(D) LPS induction of tolerance in vivo is dose dependent. Mice (n = 3–6) received various doses of E. coli O14 LPS or PBS intraperitoneally on day 0. On day 14,
isolated peritoneal macrophages were exposed to 1 mg/ml LPS for 6 hr, and medium IL-6 was measured. Error bars represent ± 1 SEM.
(E) Responses to LPS of macrophages explanted from mice primed with 10 mg LPS 8 weeks previously. In (A–E), significant differences from PBS-primed mice of
same genotype are indicated by * p < 0.05; **p < 0.01; *** p < 0.001.
(F) Recombinant AOAH prevents prolonged tolerance in Aoah / mice. Mice that had received an intravenous injection of 1 3 108 transgene equivalents of
Ad.CMV-hAOAH or Ad.CMV-eGFP 14 days previously were primed intraperitoneally with PBS or 1 mg LPS. Peritoneal macrophages were isolated 14 days later
and stimulated with LPS ex vivo. The *** denote p < 0.001 versus other groups. Data were combined from two independent experiments. The symbol designations
in (A) are used in the other panels. Here, the open circles represent LPS-primed Aoah / mice that had received Ad.CMV-hAOAH.

either 1 mg LPS or PBS intraperitoneally. When they were har- phages was FITC+ in both Aoah / and Aoah+/+ mice. The mean
vested 2 weeks later and stimulated with LPS ex vivo, macro- fluorescence intensity was similar for macrophages of both ge-
phages from LPS-primed Aoah / mice that had received notypes. To measure the uptake and deacylation of LPS by peri-
Ad.CMV-hAOAH produced 20-fold more IL-6 and TNF than did toneal cells in vivo, we used [3H/14C]LPS. Ten days after intraper-
macrophages from Aoah / mice that had received a control itoneal injection of 10 mg radiolabeled LPS, more of the LPS
virus, Ad.CMV-eGFP (Figure 3F), and their ability to produce remained associated with peritoneal cells in Aoah / mice,
several other cytokines and chemokines in response to LPS which had more macrophages (Figure 4B, left); very little of
was also restored (Table S1). Providing recombinant AOAH this LPS had been deacylated (Figure 4B, right). In contrast,
thus prevented prolonged tolerance in Aoah / animals. the [3H/14C]LPS found in Aoah+/+ cells lacked one-third of its
3
H-acyl chains, consistent with loss of the two secondary acyl
Retention of Acylated LPS by Macrophages chains that are released by AOAH (Shao et al., 2007). Collec-
Is Associated with Prolonged Tolerance In Vivo tively, these findings suggest that the macrophages from LPS-
Ten days after intraperitoneal priming there were more macro- primed Aoah / and Aoah+/+ mice contained similar amounts
phages, neutrophils, and B cells in the peritoneal washes of of LPS per cell when they were studied 10 days later; in Aoah+/+
Aoah / mice that had been primed with LPS than in the other macrophages, the LPS had lost one-third of its acyl chains,
groups (Table S2). Flow cytometric examination of peritoneal whereas in Aoah / macrophages it remained fully acylated. Im-
cells 10 days after intraperitoneal injection of a fluorescent LPS portantly, production of TNF following LPS exposure ex vivo was
preparation (FITC-LPS) revealed that the FITC-LPS was associ- inversely related to the amount of cell-associated 14C-LPS
ated almost exclusively with F4/80+ cells (macrophages) (Figure 4C). Prolonged tolerance thus correlated directly with
(Figure 4A). Approximately 60% to 70% of the recovered macro- the retention of fully acylated LPS by peritoneal macrophages.

296 Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

Prolonged Tolerance Is Immunosuppressive


To find out how prolonged in vivo tolerance influences host de-
fense, we compared the ability of Aoah / and Aoah+/+ mice to
resist a virulent E. coli O18 strain (Cross et al., 1995) two weeks
after a priming dose of LPS. Whereas no PBS-primed Aoah+/+
mice succumbed to the bacterial challenge, 90% of the LPS-
primed Aoah / mice did not survive (Figure 5A). When we
repeated the bacterial challenge and measured cfus in various
organs 24 hr later, we recovered 102- to 103-fold more cfus
from the blood of LPS-primed Aoah / mice than from mice in
the other experimental groups (Figure 5B). A similar pattern
was observed in the liver, spleen, lung, kidney and peritoneal
fluid (data not shown). In the absence of AOAH, LPS priming
thus severely impairs the ability to control E. coli infection.
The impact of prolonged tolerance was evident shortly follow-
ing the infectious challenge. One hour after challenge with 3 3
107 E. coli O18K1, LPS-primed Aoah / mice had lower plasma
TNF (Figure 5C) and IL-6 (data not shown) than did animals in
the other groups; the IL-10 response was more variable
(Figure 5D). Similar results were found when mice had been
primed with live E. coli and then challenged with the virulent
O18K1 strain (Figure S2). Animals that retained fully acylated
LPS in peritoneal macrophages thus mounted sluggish pro-in-
flammatory cytokine responses when challenged with live bac-
teria. Twenty-four hours after infection, the plasma and perito-
neal levels of TNF and IL-6 (data not shown) were directly
related to the numbers of bacteria found in these fluids (Figures
5E and 5F). Uncontrolled infection in LPS-primed Aoah / mice
thus can induce inflammatory responses that likely contribute to
lethality.

DISCUSSION

We studied the duration of endotoxin tolerance in vivo and found


strong evidence that terminating tolerance to Gram-negative
bacteria requires enzymatic inactivation of their major TLR
ligand, LPS. The inactivating enzyme, AOAH, is produced by
monocyte macrophages, neutrophils, dendritic cells (Lu et al.,
2003), and renal cortical epithelial cells (Feulner et al., 2004). It
removes the secondary acyl chains that are required for LPS
recognition by MD-2—TLR4, the LPS-signaling receptor (Mun-
ford and Varley, 2006). LPS deacylation occurs over many hours
in vivo and does not appreciably diminish the acute inflammatory
response to LPS or Gram-negative bacteria. On the other hand,
persistently acylated LPS can induce prolonged, exaggerated
antibody responses (Lu et al., 2005), hepatomegaly (Shao
Figure 4. Fully Acylated LPS Remains Macrophage-Associated in et al., 2007), and low-grade elevations in circulating levels of cer-
Primed Aoah / Mice and Induces Prolonged Reprogramming
tain cytokines (IL-10) (B. Shao, unpublished data). Here we found
(A) Flow cytometric analysis of peritoneal monocyte macrophages harvested
10 days after intraperitoneally injection of FITC-LPS, LPS, or FITC glycine.
that AOAH deficiency has a profound impact on the intensity and
(B) Peritoneal cells were harvested from Aoah+/+ and Aoah / mice that had duration of endotoxin tolerance. The LPS dose required to in-
received intraperitoneal injections of 10 mg [3H/14C]LPS 10 days earlier. Recov- duce prolonged reprogramming in animals that could not deacy-
ery of LPS (left) and its deacylation (percentage loss of 3H dpm, right) are late LPS was at least 20-fold lower than that needed in wild-type
shown. AOAH removes two of the six acyl chains. Error bar represents 1 mice (Figure 3D), and the tolerant state lasted for at least 2
SEM and n = seven to eight mice/group.
months (Figure 3E). The intensity of the tolerant state, as as-
(C) Ten days after LPS intraperitoneal injection, there was an inverse correla-
sessed by the production of TNF by explanted macrophages,
tion between the amount of TNF produced following LPS stimulation of ex-
planted macrophages ex vivo and the amount of radiolabeled LPS associated was directly related to the amount of macrophage-associated
with the explanted cells. acylated LPS (Figure 4C). Prolonged tolerance did not occur
in Aoah / animals when MD-2—TLR4 was activated by an
agonistic monoclonal antibody (Figure 1F), providing strong

Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc. 297
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

Figure 5. LPS-Primed Aoah / Mice Are


More Susceptible to E. coli Challenge
(A) Groups of mice received a priming intraperito-
neal dose of 10 mg E. coli O14 LPS or PBS. Ten to
14 days later, they were challenged intraperitone-
ally with 800 cfus E. coli O18K1. Data combined
from three independent experiments (n = 15–19)
are shown. Each survival curve is different from
the others at p % 0.01 (Mantel-Cox).
(B) In another experiment, cultures were per-
formed 24 hr after intraperitoneal inoculation of
800 cfus E. coli. The figure shows the cfus recov-
ered from cultures of blood.
(C and D) In a third experimental design, 5 3 107
cfus E. coli were injected intraperitoneally, and
plasma cytokine levels were measured 1 hr later.
At this time point, plasma TNF levels (C) were sig-
nificantly lower in primed Aoah / mice than in the
other groups, whereas plasma IL-10 was more
variable (D). Data combined from two experi-
ments, each with n = 4. Error bars = 1 SEM.
(E and F) Twenty-four hours postchallenge (800
cfus), in contrast, the concentrations of TNF in
the blood (E) correlated (F) with the number of
recovered bacteria in (B). Similar correlations
were observed between IL-6 and cfus and for
both peritoneal fluid and blood (data not shown).
* p < 0.05; **p < 0.01; *** p < 0.001.

Many previous investigators have


found that early endotoxin tolerance
(lasting hours or a few days) can boost
host defenses (Lehner et al., 2001; Freu-
denberg et al., 2001; Cross, 2002; Cavail-
lon et al., 2003; Echtenacher and Mannel,
2002). Enhanced resistance to infectious
challenge during early tolerance might
have several mechanisms: the ability of
TNF (produced in response to the priming
dose of LPS) to boost host defenses
evidence that the enzyme’s ability to inactivate LPS is essential (Echtenacher and Mannel, 2002; Cavaillon et al., 2003), a greater
to its ability to limit tolerance. influx of neutrophils into the site of bacterial inoculation (Lehner
In the categorization developed by Greisman (Greisman et al., et al., 2001), diminished neutrophil apoptosis (Feterowski et al.,
1969), early endotoxin tolerance is mediated by changes in 2001), interferon-mediated hypersensitivity to LPS (Freudenberg
cellular responsiveness to LPS (this phase was later attributed et al., 2001), or increased production of antimicrobial proteins
to macrophages (Freudenberg and Galanos, 1988), whereas (Foster et al., 2007). In contrast, other groups have found that
(late) tolerance that lasts more than a few days is caused by neu- early tolerance can diminish host defenses. In particular, toler-
tralization of the challenge LPS dose by antibodies raised in re- ance induced by intraperitoneal infection or intraportal injection
sponse to the initial LPS exposure. The latter phenomenon of LPS can diminish antibacterial resistance in the lungs of
seemed a likely explanation for prolonged tolerance in Aoah / mice (Mason et al., 1997; Deng et al., 2006), pointing to complex
mice, which react to low doses of LPS by mounting robust interactions between the systemic and local compartments (Fit-
polyclonal IgM and IgG3 responses that include antibodies to ting et al., 2004). In all of these studies, microbial challenge was
many different LPSs (Lu et al., 2005). Indeed, we also found in- administered within a few hours or days of the priming exposure.
creased numbers of B cells in the peritoneal washes of LPS- The interval between primary LPS exposure and bacterial
primed Aoah / mice (Table S1). We were thus surprised to challenge was much longer in our studies than in those previ-
find that prolonged LPS reprogramming occurred in Aoah / ously reported. Remarkably, for at least two weeks after LPS
mice that lack B cells (Figure 2) and was also present in macro- priming, AOAH-deficient animals had increased numbers of in-
phages that had been removed from the antibody-rich in vivo traperitoneal B cells and macrophages (Table S2), suggesting
environment (Figure 3). In animals that cannot deacylate LPS, that the fully acylated LPS remains stimulatory in vivo. Nonethe-
prolonged endotoxin tolerance is not mediated by antibodies less, they mounted sluggish proinflammatory (TNF, IL-6) re-
or B cells. sponses when challenged with live bacteria (Figures 4C and

298 Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

S2), and were more likely to die following intraperitoneal bacterial et al., 2005), until recently there was only indirect evidence that
challenge (Figure 4A). Moreover, macrophages explanted from endogenous phosphatases play this role. Inactivation of LPS
LPS-primed Aoah / mice produced significantly lower levels by an intestinal alkaline phosphatase has now been reported to
of many chemokines and cytokines when they were stimulated prevent gut-mucosal inflammation in zebrafish (Bates et al.,
ex vivo with LPS (Figure 3 and Table S1). The immunodeficiency 2007), which do not produce AOAH, suggesting that LPS de-
observed here seems quite similar to that described by Cross phosphorylation may be an important mechanism for inactivating
et al., who found that TLR4-dependent inflammatory responses the large amounts of LPS found in the intestine. Our findings in-
were indispensable for host defense versus virulent E. coli chal- dicate, in contrast, that enzymatic deacylation plays a distinctive
lenge (Cross et al., 1995). In the present case, it seems likely that role in inactivating LPS in extraintestinal tissues, even in animals
a dampened, sluggish initial response allowed bacteria to prolif- that produce endogenous phosphatases (Peterson and Mun-
erate to levels that then induced sustained, harmful levels of ford, 1987; Lu et al., 2005; Shao et al., 2007; Vaishnava and
proinflammatory mediators. Hooper, 2007). Most importantly, we have provided evidence
Several previous observations have suggested that persis- that disabling this potent microbial agonist is required to restore
tence of MD-2 —TLR4 agonists can prolong tolerance in vivo. effective antibacterial host defenses following Gram-negative
Whereas a single intravenous dose of LPS could induce toler- bacterial infection in vivo. It is possible that host mechanisms
ance in humans for less than 14 days (Engelhardt et al., 1991), for inactivating other microbial molecules, such as bacterial lipo-
daily administration of LPS maintained tolerance for as long as peptides and peptidoglycan fragments (Hedl et al., 2007) or viral
30 days (Greisman et al., 1963) and weekly injections maintained TLR agonists (Didierlaurent et al., 2008), may also play a role in
tolerance for as long as 42 days (Engelhardt et al., 1991). In mice, terminating infection-induced immunosuppression in animals.
which generally recover from LPS-induced tolerance within 5 to
7 days (Wysocka et al., 2001), the agonistic UT12 monoclonal EXPERIMENTAL PROCEDURES
antibody to MD-2/TLR4 induced tolerance that lasted at least 9
Mice
days (Ohta et al., 2006). These reports and the inverse correlation
Targeted disruption of mouse Aoah was accomplished as described (Lu et al.,
found here between ex vivo TNF production and macrophage-
2003). The mutation was backcrossed eight generations into the C57Bl/6 and
associated LPS (Figure 4C) suggest that persistent TLR4 activa- C3H/HeN backgrounds. All mice were kept in a specific pathogen-free barrier
tion is sufficient to perpetuate, at least in part, mechanisms that facility. mMT Aoah / double knockouts, obtained by crossing mMT with
reprogram cellular responses to LPS and several other microbial Aoah / mice, lacked serum IgM and IgG. Mouse protocols were approved
agonists. Remarkably, LPS-primed Aoah / mice may not re- by the Institutional Animal Care and Utilization Committee of The University
gain normal responsiveness to LPS. For convenience we termi- of Texas Southwestern Medical Center (Dallas, TX).

nated almost all experiments 10 to 14 days after priming, a time


Reagents
at which significant differences between Aoah / and Aoah+/+ E. coli O14 LPS was prepared by the phenol-chloroform-petroleum ether
mice were consistently found. LPS-primed Aoah / mice re- method (Galanos et al., 1969). Rc S. typhimurium LPS (3H-labeled fatty acyl
mained tolerant in vivo for at least 22 days (Figure 1), however, chains and 14C-labeled glucosamine backbone) was prepared as described
and explanted macrophages were reprogrammed when they previously (Munford and Erwin, 1992); 1 mg had approximately 150,000 dpm
3
were studied 8 weeks after the priming LPS dose (Figure 3E). H and 10,000 dpm 14C. We measured the ratio of 3H dpm to 14C dpm in
cell lysates to estimate percent deacylation (Shao et al., 2007). FITC-LPS
In other studies we have found that LPS-exposed Aoah /
was prepared as described by Tobias et al. (1995). LPS from E. coli O14
mice develop lymph node enlargement (Lu et al., 2005) and he- was resuspended (2 mg/ml) in 0.1 M borate, pH 10.5. Five micrograms radio-
patomegaly (Shao et al., 2007) that lasted at least 6 and 4 weeks, labeled LPS were added so that the concentration of the final product could be
respectively. Enzymatic deacylation may thus be required to calculated. Ten milligrams of solid FITC was then added to 2.5 ml suspension
terminate many different responses to LPS in vivo. and incubated for 3 hr at 37 C. A 10-fold excess of glycine was added to stop
We found that providing AOAH in an adenoviral vector could the reaction. The suspension was dialyzed (1000 MW cut off) against PBS until
prevent prolonged LPS-induced tolerance in Aoah / animals the dialysis buffer was no longer yellow. The FITC-LPS was then precipitated
by adding a 2-fold excess of ethanol. The pellet was washed three times with
(Figure 2F). This result raises the possibility that increasing
70% ethanol and resuspended in PBS. The degree of labeling was 0.17 FITC/
AOAH activity in vivo might hasten the recovery of host defenses LPS (mol/mol). Glycine-FITC was made by mixing glycine with FITC in PBS.
following tissue-invasive Gram-negative bacterial diseases and The solution was diluted so that its OD494 was the same as that of the FITC-
help prevent secondary infections. Enhancing AOAH activity LPS. The FITC mean fluorescence intensity (MFI) of peritoneal cells did not
might also improve pulmonary defenses in individuals whose al- change after adding trypan blue, which quenches extracellular FITC. To en-
veolar macrophages have been tolerized by the endotoxin in to- hance intracellular FITC, cells were permeabilized with 0.5% saponin (Sigma)
and then stained with Qdot 565 Goat Anti-Fluorescein Conjugate (Invitrogen).
bacco smoke (Chen et al., 2007), enhance responses to infection
Murine monoclonal anti-MD-2/TLR4 antibody, UT12 (Ohta et al., 2006), was
in patients with hepatic cirrhosis, whose circulating monocytes prepared from mouse ascites using a T-GEL MacroPAC Kit from SCIPAC (Sit-
may be tolerized by gut-derived endotoxin (Von Baehr et al., tingbourne, Kent, UK). Purity was ascertained by SDS-PAGE with Coomassie
2000), or diminish systemic immune activation in patients with blue staining. The purified mAb was approximately 500-fold more potent
human immunodeficiency virus disease (Brenchley et al., than heat-denatured mAb (95 C, 5 min) at eliciting TNF release from C56Bl/
2006). It should also be worthwhile to look for enhanced infection 6 (Tlr4+/+) murine macrophages, and it did not stimulate Tlr4 / macrophages
to release TNF (data not shown).
susceptibility related to differences in AOAH expression among
such individuals (Gao et al., 2007).
Gutted Adenoviral Vectors
Although exogenously-administered alkaline phosphatase A vector that expresses human AOAH (gAd.CMV-hAOAH) was prepared by
can evidently inactivate LPS in vivo in rodents (Beumer et al., cloning the 1.7 kb AOAH cDNA into a gutted-Ad shuttle plasmid, pACPar-
2003; Koyama et al., 2002; Poelstra et al., 1997; van Veen entCmF.CMVp-hGHTerm, as an EcoRI fragment. The product was linearized

Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc. 299
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

with PmeI and cotransformed with undigested gutted-Ad packaging plasmid Medicine, UT-Southwestern Medical Center. We thank F. L. Graham and
(pAd4) into E. coli strain BJ5183 (a gift from B. Vogelstein, Johns Hopkins B. Vogelstein for essential reagents; David Farrar, Beth Levine and Lora
School of Medicine) to allow homologous recombination (He et al., 1998), Hooper for helpful advice; and Baomei Shao for titrating the adenovirus preps.
which results in the transfer of the expression cassette and Kan marker from
the shuttle plasmid into the packaging plasmid. Kanamycin-resistant clones Received: March 6, 2008
were screened by digestion with PacI, and clones showing the expected pat- Revised: June 2, 2008
tern were verified by transformation into 293 cells followed by assaying for Accepted: June 30, 2008
AOAH activity. The finished packaging plasmid was then digested with PacI Published: September 10, 2008
to liberate the complete gutted-Ad genome, which was transformed into
116cre cells (Parks et al., 1996). High titer, purified gAd.CMV-hAOAH was REFERENCES
then prepared by the cre-lox system of Graham and coworkers (Parks et al.,
1996) (6) as modified by Palmer and Ng (Palmer and Ng, 2003). A control virus Alexopoulou, L., Holt, A.C., Medzhitov, R., and Flavell, R.A. (2001). Recogni-
that expresses eGFP (cDNA from Clontech, Palo Alto, CA) was produced in the tion of double-stranded RNA and activation of NF-kappaB by Toll- like recep-
same way. The viral preps were assayed by quantitative PCR to determine the tor 3. Nature 413, 732–738.
number of transgene equivalents per milliliter. In preliminary experiments, we Bagchi, A., Herrup, E.A., Warren, H.S., Trigilio, J., Shin, H.S., Valentine, C., and
found that sustained production of AOAH in vivo occurred from 2 to 4 weeks Hellman, J. (2007). MyD88-dependent and MyD88-independent pathways in
after intravenous injection of Ad.CMV-hAOAH. synergy, priming, and tolerance between TLR agonists. J. Immunol. 178,
1164–1171.
In Vivo and Ex Vivo Experiments
Bates, J.M., Akerlund, J., Mittge, E., and Guillemin, K. (2007). Intestinal alkaline
Mice were injected intraperitoneally with 10 mg E. coli 014 LPS in 300 ml PBS,
phosphatase detoxifies lipopolysaccharide and prevents inflammation in ze-
1 mg UT12 mAb in PBS, 1.5 3 107 E. coli LCD25 (Munford et al., 1992) or
brafish in response to the gut microbiota. Cell Host Microbe 2, 371–382.
PBS only. This E. coli inoculum contains approximately 150 ng LPS (Ingraham
et al., 1983). They were challenged on later dates with 20 mg E. coli O111 LPS in Beumer, C., Wulferink, M., Raaben, W., Fiechter, D., Brands, R., and Seinen,
300 ml PBS intraperitoneally. Tail-vein blood was obtained 1 and 4 hr after chal- W. (2003). Calf intestinal alkaline phosphatase, a novel therapeutic drug for li-
lenge. EDTA was used as anticoagulant. Live bacterial challenge was per- popolysaccharide (LPS)-mediated diseases, attenuates LPS toxicity in mice
formed using E. coli O18K1, a virulent strain provided by A. Cross (University and piglets. J. Pharmacol. Exp. Ther. 307, 737–744.
of Maryland School of Medicine). For ex vivo experiments, mice were primed Björkbacka, H., Fitzgerald, K.A., Huet, F., Li, X., Gregory, J.A., Lee, M.A.,
with E. coli 014 LPS or PBS intraperitoneally; on some occasions, two priming Ordija, C.M., Dowley, N.E., Golenbock, D.T., and Freeman, M.W. (2004). The
doses were given. Ten or 14 days after the last priming dose, mice were eutha- induction of macrophage gene expression by LPS predominantly utilizes
nized with CO2, and their peritoneal cells were flushed with 5 ml PBS containing Myd88-independent signaling cascades. Physiol. Genomics 19, 319–330.
3 mM EDTA. After adherence for 16 hr and washing with complete RPMI Brenchley, J.M., Price, D.A., Schacker, T.W., Asher, T.E., Silvestri, G., Rao, S.,
(cRPMI) medium (Lu et al., 2005), the cells were treated with 1 mg/ml of E. coli Kazzaz, Z., Bornstein, E., Lambotte, O., Altmann, D., et al. (2006). Microbial
O111 LPS (Sigma), 40 mg/ml of killed Micrococcus luteus cells (Sigma), or translocation is a cause of systemic immune activation in chronic HIV infection.
10 mg/ml of poly I:C (Sigma) for 6 hr. The media were harvested for cytokine as- Nat. Med. 12, 1365–1371.
says, and the cells were washed prior to lysis in PBS 0.1% Triton X-100 to mea-
Cavaillon, J.M., Adrie, C., Fitting, C., and Adib-Conquy, M. (2003). Endotoxin
sure cell protein (Biorad). In results not shown, the TNF response to polyI:C was
tolerance: is there a clinical relevance? J. Endotoxin Res. 9, 101–107.
very low in all of the study groups, indicating that the preparation used here was
endotoxin free, and Micrococcus luteus cells induced similar production of TNF Chen, H., Cowan, M.J., Hasday, J.D., Vogel, S.N., and Medvedev, A.E. (2007).
by Tlr4+/+ and Tlr4 / murine macrophages, as expected for a TLR2 agonist. Tobacco smoking inhibits expression of proinflammatory cytokines and
activation of IL-1R-associated kinase, p38, and NF-{kappa}B in Alveolar
macrophages stimulated with TLR2 and TLR4 agonists. J. Immunol. 179,
Assays
6097–6106.
The RANTES, TGFb, and MIP-1a ELISA Kits were from R&D Systems. The IL-1b
ELISA Kit from eBioscience and the Griess Reagent Kit were from Invitrogen. Cross, A.S. (2002). Endotoxin tolerance–current concepts in historical per-
Other cytokines and chemokines were measured using ELISA Kits from Bec- spective. J. Endotoxin Res. 8, 83–98.
ton-Dickenson or a Searchlight Array (Thermo Scientific Life Science Research Cross, A., Asher, L., Seguin, M., Yuan, L., Kelly, N., Hammack, C., Sadoff, J.,
Products, Rockford, IL). Flow cytometry and data analysis were performed as and Gemski, P., Jr. (1995). The importance of a lipopolysaccharide-initiated,
described (Lu et al., 2005). Fc binding was blocked with 0.5 mg/ml cytokine-mediated host defense mechanism in mice against extraintestinally
of normal mouse IgG (Caltag). Cells were then incubated with antibodies for invasive Escherichia coli. J. Clin. Invest. 96, 676–686.
1 hr on ice, washed, and analyzed using FACScan (Becton-Dickenson) with Deng, J.C., Cheng, G., Newstead, M.W., Zeng, X., Kobayashi, K., Flavell, R.A.,
Flow Jo Software (TreeStar, Inc.). Anti-mouse F4/80-FITC (Clone CI:A3-1) and Standiford, T.J. (2006). Sepsis-induced suppression of lung innate immu-
was from Serotec; Anti-mouse F4/80 (Biotin and PE) (Clone BM8) was from nity is mediated by IRAK-M. J. Clin. Invest. 116, 2532–2542.
Caltag. CD11b Clone M1/70, Gr-1 Clone RB6-8C5, B220 Clone RA3-6B2,
Didierlaurent, A., Goulding, J., Patel, S., Snelgrove, R., Low, L., Bebien, M.,
CD5 Clone 53-7.3, CD3 Clone 145-2C11, NK1.1 Clone PK136, and CD11c
Lawrence, T., van Rijt, L.S., Lambrecht, B.N., Sirard, J.C., and Hussell, T.
Clone HL3 were all from Becton-Dickenson.
(2008). Sustained desensitization to bacterial Toll-like receptor ligands after
resolution of respiratory influenza infection. J. Exp. Med. 205, 323–329.
Statistics
Unpaired Student’s t test (two-tailed) was used for comparisons between Dobrovolskaia, M.A., Medvedev, A.E., Thomas, K.E., Cuesta, N., Toshchakov,
groups. Log-rank (Mantel-Cox) test was used for survival-curve analysis. V., Ren, T., Cody, M.J., Michalek, S.M., Rice, N.R., and Vogel, S.N. (2003).
Induction of in vitro reprogramming by Toll-like receptor (TLR)2 and TLR4
agonists in murine macrophages: effects of TLR ‘‘homotolerance’’ versus
SUPPLEMENTAL DATA
‘‘heterotolerance’’ on NF-kappa B signaling pathway components. J. Immunol.
170, 508–519.
Supplemental Data include two tables and two figures and can be found online
at http://www.cellhostandmicrobe.com/cgi/content/full/4/3/293/DC1/. Echtenacher, B., and Mannel, D.N. (2002). Requirement of TNF and TNF
receptor type 2 for LPS-induced protection from lethal septic peritonitis. J.
ACKNOWLEDGMENTS Endotoxin Res. 8, 365–369.
Engelhardt, R., Mackensen, A., and Galanos, C. (1991). Phase I Trial of Intra-
This work was supported by grant AI18188 from the National Institute of Al- venously Administered Endotoxin (Salmonella abortus equi) in Cancer
lergy and Infectious Diseases and the Jan and Henri Bromberg Chair in Internal Patients. Cancer Res. 51, 2524–2530.

300 Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

Faist, E., Mewes, A., Strasser, T., Walz, A., Alkan, S., Baker, C., Ertel, W., and Lu, M., Zhang, M., Kitchens, R.L., Fosmire, S., Takashima, A., and Munford,
Heberer, G. (1988). Alteration of monocyte function following major injury. R.S. (2003). Stimulus-dependent deacylation of bacterial lipopolysaccharide
Arch. Surg. 123, 287–292. by dendritic cells. J. Exp. Med. 197, 1745–1754.
Feterowski, C., Weighardt, H., Emmanuilidis, K., Hartung, T., and Holzmann, Lu, M., Zhang, M., Takashima, A., Weiss, J., Apicella, M.A., Li, X.H., Yuan, D.,
B. (2001). Immune protection against septic peritonitis in endotoxin-primed and Munford, R.S. (2005). Lipopolysaccharide deacylation by an endogenous
mice is related to reduced neutrophil apoptosis. Eur. J. Immunol. 31, 1268– lipase controls innate antibody responses to Gram-negative bacteria. Nat. Im-
1277. munol. 6, 989–994.
Feulner, J.A., Lu, M., Shelton, J.M., Zhang, M., Richardson, J.A., and Munford, Mason, C.M., Dobard, E., Summer, W.R., and Nelson, S. (1997). Intraportal li-
R.S. (2004). Identification of acyloxyacyl hydrolase, a lipopolysaccharide-de- popolysaccharide suppresses pulmonary antibacterial defense mechanisms.
toxifying enzyme, in the murine urinary tract. Infect. Immun. 72, 3171–3178. J. Infect. Dis. 176, 1293–1302.
Fitting, C., Dhawan, S., and Cavaillon, J. (2004). Compartmentalization of Tol- Mata-Haro, V., Cekic, C., Martin, M., Chilton, P.M., Casella, C.R., and Mitchell,
erance to Endotoxin. J. Infect. Dis. 189, 1295–1303. T.C. (2007). The Vaccine Adjuvant Monophosphoryl Lipid A as a TRIF-Biased
Foster, S.L., Hargreaves, D.C., and Medzhitov, R. (2007). Gene-specific con- Agonist of TLR4. Science 316, 1628–1632.
trol of inflammation by TLR-induced chromatin modifications. Nature 447,
McCabe, W.R. (1963). Endotoxin tolerance. II. Its occurrence in patients with
972–978.
pyelonephritis. J. Clin. Invest. 42, 618–625.
Freudenberg, M.A., and Galanos, C. (1988). Induction of tolerance to lipopoly-
McCall, C.E., and Yoza, B.K. (2007). Gene silencing in severe systemic inflam-
saccharide (LPS)-D-galactosamine lethality by pretreatment with LPS is medi-
mation. Am. J. Respir. Crit. Care Med. 175, 763–767.
ated by macrophages. Infect. Immun. 56, 1352–1357.
McCall, C.E., Grosso-Wilmoth, L.M., LaRue, K., Guzman, R.N., and Cousart,
Freudenberg, M.A., Merlin, T., Gumenscheimer, M., Kalis, C., Landmann, R.,
S.L. (1993). Tolerance to endotoxin-induced expression of the interleukin- 1b
and Galanos, C. (2001). Role of lipopolysaccharide susceptibility in the innate
gene in blood neutrophils of humans with the sepsis syndrome. J. Clin. Invest.
immune response to Salmonella typhimurium infection: LPS, a primary target
91, 853–861.
for recognition of Gram-negative bacteria. Microbes Infect. 3, 1213–1222.
Galanos, C., Luderitz, O., and Westphal, O. (1969). A new method for the ex- Medvedev, A.E., Sabroe, I., Hasday, J.D., and Vogel, S.N. (2006). Tolerance to
traction of R lipopolysaccharides. Eur. J. Biochem. 9, 245–249. microbial TLR ligands: molecular mechanisms and relevance to disease. J. En-
dotoxin Res. 12, 133–150.
Gao, L., Tsai, Y.J., Grigoryev, D.N., and Barnes, K.C. (2007). Host defense
genes in asthma and sepsis and the role of the environment. Curr. Opin. Allergy Munford, R.S. (2008). Sensing gram-negative bacterial lipopolysaccharides:
Clin. Immunol. 7, 459–467. a human disease determinant? Infect. Immun. 76, 454–465.
Greisman, S.E., Hornick, R.B., Carozza, F.A., Jr., and Woodward, T.E. (1963). Munford, R.S., and Hall, C.L. (1986). Detoxification of bacterial lipopolysac-
The role of endotoxin during typhoid fever and tularemia in man. I. Acquisition charides (endotoxins) by a human neutrophil enzyme. Science 234, 203–205.
of tolerance to endotoxin. J. Clin. Invest. 42, 1064–1075. Munford, R.S., and Erwin, A.L. (1992). Eucaryotic lipopolysaccharide deacylat-
Greisman, S.E., Young, E.J., and Carozza, F.A., Jr. (1969). Mechanisms of ing enzyme. Methods Enzymol. 209, 485–492.
Endotoxin Tolerance: V. Specificity of the early and late phases of pyrogenic Munford, R.S., and Varley, A.W. (2006). Shield as signal: lipopolysaccharides
tolerance. J. Immunol. 103, 1223–1236. and the evolution of immunity to Gram-negative bacteria. PLoS Pathog. 2, e67.
He, T.-C., Zhou, S., da Costa, L.T., Yu, J., Kinzler, K.W., and Vogelstein, B.
Munford, R.S., DeVeaux, L.C., Cronan, J.E., Jr., and Rick, P.D. (1992). Biosyn-
(1998). A simplified system for generating recombinant adenoviruses. Proc.
thetic radiolabeling of bacterial lipopolysaccharide to high specific activity. J.
Natl. Acad. Sci. USA 95, 2509–2514.
Immunol. Methods 148, 115–120.
Hedl, M., Li, J., Cho, J.H., and Abraham, C. (2007). Chronic stimulation of Nod2
Munoz, C., Carlet, J., Fitting, C., Misset, B., Bieriot, J.-P., and Cavaillon, J.-M.
mediates tolerance to bacterial products. Proc. Natl. Acad. Sci. USA 104,
(1991). Dysregulation of in vitro cytokine production by monocytes during sep-
19440–19445.
sis. J. Clin. Invest. 88, 1747–1754.
Helminen, M., and Vesikari, T. (1990a). Increased interleukin-1 (IL-1) produc-
Ohta, S., Bahrun, U., Shimazu, R., Matsushita, H., Fukudome, K., and Kimoto,
tion from LPS-stimulated peripheral blood monocytes in children with febrile
M. (2006). Induction of Long-Term Lipopolysaccharide Tolerance by an Ago-
convulsions. Acta Paediatr. Scand. 79, 810–816.
nistic Monoclonal Antibody to the Toll-Like Receptor 4/MD-2 Complex. Clin.
Helminen, M., and Vesikari, T. (1990b). Interleukin-1 production in bacterial
Vaccine Immunol. 13, 1131–1136.
meningitis. Scand. J. Infect. Dis. 22, 105–108.
Palmer, D., and Ng, P. (2003). Improved system for helper-dependent adeno-
Henricson, B.E., Manthey, C.L., Yu Perera, P., Hamilton, T.A., and Vogel, S.N.
viral vector production. Mol. Ther. 8, 846–852.
(1993). Dissociation of lipopolysaccharide (LPS)-inducible gene expression in
murine macrophages pretreated with smooth LPS versus monophosphoryl Parks, R.J., Chen, L., Anton, M., Sankar, U., Rudnicki, M.A., and Graham, F.L.
lipid A. Infect. Immun. 61, 2325–2333. (1996). A helper-dependent adenovirus vector system: Removal of helper virus
by Cre-mediated excision of the viral packaging signal. Proc. Natl. Acad. Sci.
Ingraham, J.L., Maaloe, O., and Neidhardt, F.C. (1983). Growth of the Bacterial
USA 93, 13565–13570.
Cell (Sunderland, MA: Sinauers Associates, Inc.).
Kelly, J.L., O’Sullivan, C., O’Riordain, M., O’Riordain, D., Lyons, A., Doherty, J., Peterson, A.A., and Munford, R.S. (1987). Dephosphorylation of the lipid A
Mannick, J.A., and Rodrick, M.L. (1997). Is circulating endotoxin the trigger for moiety of Escherichia coli lipopolysaccharide by mouse macrophages. Infect.
the systemic inflammatory response syndrome seen after injury. Ann. Surg. Immun. 55, 974–978.
225, 530–541. Poelstra, K., Bakker, W.W., Klok, P.A., Hardonk, M.J., and Meijer, D.K.F.
Kitamura, D., Roes, J., Kuhn, R., and Rajewsky, K. (1991). A B cell-deficient (1997). A physiologic function for alkaline phosphatase: Endotoxin detoxifica-
mouse by targeted disruption of the membrane exon of the immunoglobulin tion. Lab. Invest. 76, 319–327.
mu chain gene. Nature 350, 423–426. Sato, S., Takeuchi, O., Fujita, T., Tomizawa, H., Takeda, K., and Akira, S.
Koyama, I., Matsunaga, T., Harada, T., Hokari, S., and Komoda, T. (2002). Al- (2002). A variety of microbial components induce tolerance to lipopolysaccha-
kaline phosphatases reduce toxicity of lipopolysaccharides in vivo and in vitro ride by differentially affecting MyD88-dependent and - independent pathways.
through dephosphorylation. Clin. Biochem. 35, 455–461. Int. Immunol. 14, 783–791.
Lehner, M.D., Ittner, J., Bundschuh, D.S., Van Rooijen, N., Wendel, A., and Shao, B., Lu, M., Katz, S.C., Varley, A.W., Hardwick, J., Rogers, T.E., Ojogun,
Hartung, T. (2001). Improved innate immunity of endotoxin-tolerant mice in- N., Rockey, D.C., DeMatteo, R.P., and Munford, R.S. (2007). A Host Lipase De-
creases resistance to Salmonella enterica serovar typhimurium infection de- toxifies Bacterial Lipopolysaccharides in the Liver and Spleen. J. Biol. Chem.
spite attenuated cytokine response. Infect. Immun. 69, 463–471. 282, 13726–13735.

Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc. 301
Cell Host & Microbe
LPS Deacylation Limits Microbial Tolerance In Vivo

Shnyra, A., Brewington, R., Alipio, A., Amura, C., and Morrison, D.C. (1998). ceptor antagonist, and soluble tumour necrosis factor receptors as well as
Reprogramming of lipopolysaccharide-primed macrophages is controlled by effector cell desensitisation. Gut 47, 281–287.
a counterbalanced production of IL-10 and IL-12. J. Immunol. 160, 3729–
Wang, J.H., Doyle, M., Manning, B.J., Blankson, S., Wu, Q.D., Power, C., Ca-
3736.
hill, R., and Redmond, H.P. (2003). Bacterial lipoprotein induces endotoxin-in-
Somerville, J.E., Jr., Cassiano, L., Bainbridge, B., Cunningham, M.D., and Dar- dependent tolerance to septic shock. J. Immunol. 170, 14–18.
veau, R.P. (1996). A novel Escherichia coli lipid A mutant that produces an anti-
Weighardt, H., Heidecke, C.D., Emmanuilidis, K., Maier, S., Bartels, H., Sie-
inflammatory lipopolysaccharide. J. Clin. Invest. 97, 359–365.
wert, J.R., and Holzmann, B. (2000). Sepsis after major visceral surgery is as-
Tobias, P.S., Soldau, K., Gegner, J.A., Mintz, D., and Ulevitch, R.J. (1995). Li-
sociated with sustained and interferon-gamma-resistant defects of monocyte
popolysaccharide binding protein-mediated complexation of lipopolysaccha-
cytokine production. Surgery 127, 309–315.
ride with soluble CD14. J. Biol. Chem. 270, 10482–10488.
West, M.A., and Heagy, W. (2002). Endotoxin tolerance: A review. Crit. Care
Vaishnava, S., and Hooper, L.V. (2007). Alkaline phosphatase: keeping the
Med. 30, S64–S73.
peace at the gut epithelial surface. Cell Host Microbe 2, 365–367.
van Veen, S.Q., van Vliet, A.K., Wulferink, M., Brands, R., Boermeester, M.A., Wysocka, M., Robertson, S., Riemann, H., Caamano, J., Hunter, C., Mackie-
and van Gulik, T.M. (2005). Bovine Intestinal Alkaline Phosphatase Attenuates wicz, A., Montaner, L.J., Trinchieri, G., and Karp, C.L. (2001). IL-12 suppres-
the Inflammatory Response in Secondary Peritonitis in Mice. Infect. Immun. sion during experimental endotoxin tolerance: Dendritic cell loss and macro-
73, 4309–4314. phage hyporesponsiveness. J. Immunol. 166, 7504–7513.
Von Baehr, V., Döcke, W.D., Plauth, M., Liebenthal, C., Küpferling, S., Lochs, Wysocka, M., Montaner, L.J., and Karp, C.L. (2005). Flt3 ligand treatment
H., Baumgarten, R., and Volk, H.D. (2000). Mechanisms of endotoxin tolerance reverses endotoxin tolerance-related immunoparalysis. J. Immunol. 174,
in patients with alcoholic liver cirrhosis: role of interleukin 10, interleukin 1 re- 7398–7402.

302 Cell Host & Microbe 4, 293–302, September 11, 2008 ª2008 Elsevier Inc.

You might also like