You are on page 1of 12

CCR1 and CC Chemokine Ligand 5

Interactions Exacerbate Innate Immune


Responses during Sepsis
This information is current as Traci L. Ness, Kristin J. Carpenter, Jillian L. Ewing, Craig J.
of October 26, 2022. Gerard, Cory M. Hogaboam and Steven L. Kunkel
J Immunol 2004; 173:6938-6948; ;
doi: 10.4049/jimmunol.173.11.6938
http://www.jimmunol.org/content/173/11/6938

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


References This article cites 44 articles, 23 of which you can access for free at:
http://www.jimmunol.org/content/173/11/6938.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2004 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

CCR1 and CC Chemokine Ligand 5 Interactions Exacerbate


Innate Immune Responses during Sepsis1

Traci L. Ness,* Kristin J. Carpenter,* Jillian L. Ewing,* Craig J. Gerard,†


Cory M. Hogaboam,2* and Steven L. Kunkel*
CCR1 has previously been shown to play important roles in leukocyte trafficking, pathogen clearance, and the type 1/type
2 cytokine balance, although very little is known about its role in the host response during sepsis. In a cecal ligation and
puncture model of septic peritonitis, CCR1-deficient (CCR1ⴚ/ⴚ) mice were significantly protected from the lethal effects of
sepsis when compared with wild-type (WT) controls. The peritoneal and systemic cytokine profile in CCR1ⴚ/ⴚ mice was
characterized by a robust, but short-lived and regulated antibacterial response. CCR1 expression was not required for leu-
kocyte recruitment, suggesting critical differences extant in the activation of WT and CCR1ⴚ/ⴚ resident or recruited peri-
toneal cells during sepsis. Peritoneal macrophages isolated from naive CCR1ⴚ/ⴚ mice clearly demonstrated enhanced

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


cytokine/chemokine generation and antibacterial responses compared with similarly treated WT macrophages. CCR1 and
CCL5 interactions markedly altered the inflammatory response in vivo and in vitro. Administration of CCL5 increased
sepsis-induced lethality in WT mice, whereas neutralization of CCL5 improved survival. CCL5 acted in a CCR1-dependent
manner to augment production of IFN-␥ and MIP-2 to damaging levels. These data illustrate that the interaction between
CCR1 and CCL5 modulates the innate immune response during sepsis, and both represent potential targets for therapeutic
intervention. The Journal of Immunology, 2004, 173: 6938 – 6948.

S epsis is the most common cause of death in noncoronary mune response to sepsis (7, 8). Although differences in
critical care units in the U.S. with ⬎751,000 cases per year chemokine receptor expression in septic patients have been re-
at an estimated overall cost of $16.7 billion (increasing by ported (9), the significance of these differences is not under-
1.5% per year) (1). Due to the paucity of effective therapeutic stood. Almost nothing is known about the role these receptors
treatments (2), the care of septic patients is predominantly sup- play in the septic response (10, 11).
portive and mortality rates remain high. Although recombinant ac- CCR1 is expressed by a broad spectrum of leukocytes, including
tivated protein C has shown some promise (3), investigators con- neutrophils, monocytes, eosinophils, and lymphocytes (12). Stud-
tinue to search for novel therapies. ies of CCR1-deficient (CCR1⫺/⫺) mice have implicated CCR1 in
During a normal infection, the immune system of an immuno- the modulation of leukocyte trafficking (13), parasite and viral
competent host works to contain and destroy the pathogen. A sep- clearance (14, 15), and the balance of type 1 and type 2 cytokines
tic response occurs when a pathogen circumvents the innate and (16); however, little is known about its role in the innate immune
acquired immune defenses, resulting in systemic spread of the in- response during sepsis and multiorgan failure. In vitro, CCR1 has
fection. In a continued attempt to eliminate the infection, the host been shown to bind several ligands, including CCL3, CCL5–9,
enhances production of several proinflammatory cytokines and CCL14 –16, and CCL23, although CCL3, CCL5, and CCL6 are the
chemokines that act to increase the infiltration and activation of major agonists identified in vivo (17, 18). Serum levels of CCL3
inflammatory leukocytes. These factors have been shown to play a and CCL5 are elevated in septic patients (19), and peritoneal CCL3
significant role in the resulting tissue damage preceding sepsis- (20) and CCL6 (7) concentrations are increased in the cecal liga-
associated multiple organ failure (4, 5) and may serve as potential tion and puncture (CLP) mouse model of sepsis. Both CCL3 (21)
targets for immunotherapy. and CCL6 (7) have been demonstrated to play protective roles
Chemokines are a family of small, primarily secreted proteins against sepsis-induced lethality and injury in a murine CLP model,
that are responsible for modulating multiple aspects of inflam- while the role of CCL5 has yet to be explored.
matory responses and host defense (6). Many studies have dem- The purpose of this study was to investigate the role of CCR1 in the
onstrated several of these to be key mediators in the host im- innate immune response to experimental sepsis. CCR1⫺/⫺ mice were
significantly protected against CLP-induced lethality. Although
CCR1 deficiency had no effect on the inflammatory cell recruitment
*Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
48109; and †Children’s Hospital, Harvard Medical School, Boston, MA 02115
to the peritoneal cavity, loss of the receptor promoted accelerated
cytokine expression and enhanced macrophage activity, both of
Received for publication June 15, 2004. Accepted for publication September
22, 2004. which contributed to a more efficient and regulated antibacterial
The costs of publication of this article were defrayed in part by the payment of page response. CCL5 was identified as a key modulator of the host
charges. This article must therefore be hereby marked advertisement in accordance response that acted in a CCR1-dependent manner to trigger the
with 18 U.S.C. Section 1734 solely to indicate this fact. unregulated, exaggerated expression of proinflammatory cyto-
1
This work was supported by Grants HL031237 and P50HL074024 from the Na- kines, resulting in increased injury and mortality following sepsis.
tional Institutes of Health to S.L.K.
2
Address correspondence and reprint requests to Dr. Cory M. Hogaboam, Depart-
ment of Pathology, University of Michigan Medical School, 1301 Catherine Road,
3
Room 5214, Medical Sciences I, Ann Arbor, MI 48109-0602. E-mail address: Abbreviations used in this paper: CLP, cecal ligation and puncture; TSA, thymic-
hogaboam@med.umich.edu shared Ag; WT, wild type.

Copyright © 2004 by The American Association of Immunologists, Inc. 0022-1767/04/$02.00


The Journal of Immunology 6939

Materials and Methods of 300 cells from multiple high-powered fields, was multiplied by the total
peritoneal cell count to determine the number for each cell type. Three
Mice
independent experiments (three to five mice per group) showed similar
Specific pathogen-free wild-type (WT) BALB/c mice (6 – 8 wk of age) results, and data were pooled.
were purchased from The Jackson Laboratory (Bar Harbor, ME). CCR1-
deficient (CCR1⫺/⫺) mice were generated, as previously described, and Measurement of cytokines and chemokines by ELISA
were backcrossed onto a BALB/c genetic background (22). Mice were bred
Concentrations of murine TNF-␣, IFN-␥, IL-12 (p70), IL-10, MIP-2, KC,
and housed in the animal care facility (University Laboratory of Animal
CCL2, CCL3, CCL5, CCL6, CCL17, CXCL9, and CXCL10 were mea-
Medicine) at the University of Michigan. The Animal Use Committee at
sured in cell-free peritoneal lavage fluid, serum, and cell culture superna-
the University of Michigan approved all experimental procedures
tants using a standardized sandwich ELISA previously described in detail
involving mice.
(25). Briefly, flat-bottom 96-well microtiter plates (Nunc, Roskilde, Den-
Cecal ligation and puncture mark) were coated overnight at 4°C with mAb (for IL-10) or affinity-
purified polyclonal Abs for the specific cytokine of interest (R&D Systems,
CLP was used to induce acute septic peritonitis, as previously described Rochester, MN). Specific Ab for capture and detection of KC was obtained
(23). Mice were anesthetized with a combination of 2.25 mg of ketamine from PeproTech. Plates were coated with 0.4 ␮g/ml (KC), 0.5 ␮g/ml
HCl (Abbott Laboratories, Chicago, IL) and 150 ␮g of xylazine (Lloyd (IFN-␥, IL-12, CCL2, and CCL5), or 1.0 ␮g/ml (TNF-␣, IL-10, MIP-2,
Laboratories, Shenandoah, IA) administered i.p. A 1-cm incision was made CCL3, CCL6, CCL17, CXCL9, and CXCL10) appropriate capture Abs.
to the lower left abdomen of the mouse, and the cecum was exposed. The Plates were washed with PBS-Tween 20 (0.05%) and blocked with 2%
cecum was ligated distally with 3.0 silk suture and punctured through and BSA in PBS for 90 min at 37°C. Plates were rinsed four times, and samples
through with a 21- or 26-gauge needle. The cecum was returned to the were loaded and incubated at 37°C for 1 h. After washing, a biotinylated
peritoneal cavity, and surgical staples were used to close the incision. Mice secondary polyclonal Ab specific for the cytokine being measured (R&D
immediately received 1 ml of saline s.c. for fluid resuscitation and were Systems; PeproTech for KC) was added at 0.5 ␮g/ml (or 0.25 ␮g/ml for
warmed on a heating pad to facilitate their revival from the anesthetic. CCL5) for 30 min at 37°C. The plate was washed, and streptavidin-per-

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


oxidase conjugate (Bio-Rad, Richmond, CA) was added to the wells for 30
Experimental protocols min at 37°C. This was followed by another set of washes before the ad-
dition of a chromogenic substrate (Bio-Rad). After full development oc-
The first set of survival studies was performed to determine the effect of the
curred, the reaction was stopped and the plate was read in an ELISA plate
presence of CCR1 on survival following induction of acute septic perito-
reader at 490 nm. Recombinant murine cytokines/chemokines (R&D Sys-
nitis in female and male mice. Due to differential susceptibility, female
tems) were used to generate standard curves, and concentrations were ex-
mice were subjected to 21-gauge CLP, while 26-gauge CLP was performed
pressed as ng or pg/ml. Limits of detection were ⬃50 pg/ml. ELISA spec-
on male mice. Survival of CLP groups (n ⫽ 8 –10 mice per group) was
ificity was confirmed for each assay. Experimental groups consisted of
monitored for 7 days following surgery. Duplicate survival studies for the
three to five mice or triplicate samples in vitro. ELISAs were run on sam-
female mice had similar results, and, therefore, the data were pooled. For
ples from three independent experiments, and representative data are
the remaining in vivo studies, female mice were used in the 21-gauge CLP
shown.
model, while peritoneal cells were isolated from both male and female
mice for in vitro analyses, as described later.
A second set of survival studies focused on the role of exogenous and
endogenous CCL5 (RANTES) in the context of sepsis induced by 21-
gauge CLP. Two hours following CLP, 1 ␮g of murine rCCL5 (PeproTech,
Rocky Hill, NJ) or 0.1 ml of saline was administered i.p. (n ⫽ 5–10 per
group). Endogenous CCL5 was blocked in mice using a previously de-
scribed neutralizing goat polyclonal Ab against CCL5 (24) kindly provided
by R. Strieter (University of California, Los Angeles, CA). CCL5-specific
IgG was purified from antiserum using a protein G column (Pierce, Rock-
ford, IL). Mice were pretreated with 0.1 ml containing 0.5 mg of CCL5 IgG
or control goat IgG (Sigma-Aldrich, St. Louis, MO) i.p. 2 h before CLP and
every 2 days following surgery. In these studies, survival was followed for
4 days after surgery. CCL5 administration (three studies) and neutralization
(two studies) experiments (n ⫽ 5–10 per group) produced similar survival
curves, and, therefore, data were pooled for each treatment group. Female
CCR1⫺/⫺ mice were similarly treated with either saline or 1 ␮g of rCCL5
i.p. 2 h after 21-gauge CLP (n ⫽ 9 per group), after which survival was
monitored for 4 days.
In other studies, naive and CLP mice (4, 8, and 24 h after surgery; n ⫽
3– 6 per group) were anesthetized and bled. The mice were euthanized, and
peritoneal lavages were performed with 2 ml of sterile saline. Lavage fluid
was collected and saved for bacteria, cell, and protein analyses.

Determination of CFU
Blood samples obtained for the purpose of determining CFU were imme-
diately mixed with EDTA (final 4 mM) to prevent coagulation. Peritoneal
lavage fluid and EDTA-treated blood from 8- or 24-h post-CLP were
placed on ice and serially diluted in sterile saline. A 10-␮l aliquot of each
dilution was spread on thymic-shared Ag (TSA) agar plates (Difco, Detroit,
MI) and incubated at 37°C overnight. Colonies were counted and ex-
pressed as CFU/10 ␮l. Groups contained four to six mice, and the exper-
iment was repeated on one to four different occasions. Results were similar
for each experiment and were subsequently pooled. The mean for each
group was calculated and indicated by a horizontal bar.

Peritoneal leukocyte counts and cell differentials FIGURE 1. CCR1-deficient mice were less susceptible to CLP-induced
lethality. A, Female WT BALB/c (CCR1⫹/⫹) and CCR1⫺/⫺ mice were
The total number of peritoneal leukocytes per lavage sample was deter-
subjected to 21-gauge CLP. The graph represents pooled survival data from
mined by diluting 10 ␮l of lavage fluid with trypan blue and counting in a
hemocytometer. The total was expressed as leukocytes ⫻ 106 per cavity. duplicate studies showing similar results. B, Due to their increased sus-
Differential cell analyses were performed on Diff-Quik-stained cytospin ceptibility to sepsis, 26-gauge CLP was used to induce a less severe form
preparations (Dade Behring, Düdingen, Switzerland) from peritoneal la- of sepsis in the male WT and CCR1⫺/⫺ mice. Experiments contained 8 –10
vage fluid. The percentage for each leukocyte population, based on a count mice per group, and survival was followed for 7 days after surgery.
6940 CCR1 IN SEPSIS

Peritoneal macrophage isolation remove extracellular bacteria. The cells were lysed with 0.5 ml of sterile
0.5% Triton X-100 (Sigma-Aldrich). Serial 10-fold dilutions of the lysates
Naive mice were euthanized and subjected to peritoneal lavages with 10 ml were plated on TSA agar plates, and CFU were enumerated, as described
of sterile saline containing 5 mM EDTA. Lavages were pooled for mice in above.
the same group (i.e., WT or CCR1⫺/⫺). RBC were lysed in ammonium
chloride buffer (150 mM NH4Cl, 10 mM NaHCO3, 1 mM EDTA-tetraso-
dium salt), and the remaining cells were thoroughly washed with saline. Nitrite production
Cells were counted and subjected to Diff-Quik staining, as described above,
to determine the number of peritoneal macrophages. Cells were resus- NO, one of the major antimicrobial effector molecules generated by mac-
pended in complete DMEM (BioWhittaker, Walkersville, MD) containing rophages, is difficult to measure directly due to its very short t1/2. In vitro,
concentrations of nitrite (NO2⫺), a stable, oxidative end product of NO, are
5% FCS, 2 mM L-glutamine, 100 U/ml penicillin, and 100 U/ml strepto-
assessed from the medium of activated macrophages as an indirect indi-
mycin. Cells were plated in plastic plates and incubated 1–2 h at 37°C in
cator of NO production by these cells (26). Naive macrophages were plated
5% CO2. Nonadherent cells were removed, and adherent cells were washed
at 5 ⫻ 105/well of a 96-well cell culture plate (Corning Glass) and rested
with complete DMEM. Cells were either treated immediately or rested
overnight. Fresh DMEM (complete with 5% FCS) was added to each well,
overnight, depending on the assay.
and cells were treated with medium alone, 20 U/ml IFN-␥ (PeproTech),
Phagocytosis assay 100 ng/ml LPS O55:B5 (Sigma-Aldrich), or both IFN-␥ and LPS (n ⫽ 3– 6
per treatment group). After 48 h at 37°C in 5% CO2, 50 ␮l of cell-free
Escherichia coli strain O86a:K61 (American Type Culture Collection, Ma- supernatants was transferred to a flat-bottom 96-well plate and treated with
nassas, VA) was grown to mid-log phase in tryptic soy broth (Difco, De- 100 ␮l of 0.5% sulfanilamide (Sigma-Aldrich) and 0.05% naphthylethyl-
troit, MI) and used to infect freshly isolated naive macrophages (106 mac- enediamine dihydrochloride (Sigma-Aldrich) in 2.5% phosphoric acid
rophages per well) in 24-well cell culture plates (Corning Glass, Corning, (H3PO4). The absorbance was read at 550 nm in a microplate reader. A
NY). Cells were infected in antibiotic-free DMEM containing 5% FCS and standard curve was generated using known concentrations of sodium nitrite
2 mM L-glutamine at a 1:1 ratio of macrophages to bacteria. After 1 h at (NaNO2; Sigma-Aldrich) in DMEM. Similar results were shown in at least

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


37°C in 5% CO2, cells were washed four to five times with medium to three independent experiments. In vivo, NO2⫺ can be further oxidized to

FIGURE 2. CCR1⫺/⫺ mice had significantly en-


hanced bacterial clearance and reduced bacteremia after
CLP. At 8 or 24 h after CLP, mice (n ⫽ 3– 6 per group)
were bled and subjected to 2-ml peritoneal washes with
sterile saline. Serial 10-fold dilutions of peritoneal la-
vage fluid (A) and EDTA-treated blood (B) were spread
on TSA plates and incubated overnight at 37°C. Levels
of bacteria were expressed as CFU per 10 ␮l. The
graphs depict data pooled from one to four independent
studies showing similar results. The horizontal bar in-
dicates the mean for each group. C, Peritoneal lavage
fluid was collected from WT (f) and CCR1⫺/⫺ (䡺)
mice at 0, 4, 8, or 24 h post-CLP (n ⫽ 3–5 per group).
Nitrate (NO3⫺) was converted into nitrite (NO2⫺), and
then total nitrite concentration was assessed. The graph
depicts data from three pooled experiments. ⴱ, p ⬍ 0.05
compared with WT-CLP mice.
The Journal of Immunology 6941

FIGURE 3. CCR1 had no significant effect on leu-


kocyte recruitment to the peritoneal cavity after CLP.
Mice were euthanized at 0, 4, 8, or 24 h post-CLP, and
peritoneal lavages (2 ml) were collected (n ⫽ 4 – 6 per
group). A, The total number of peritoneal cells was
counted for each individual mouse. Data were collected
from three independent experiments. B, Differential counts
of mononuclear cell and neutrophil populations were per-
formed from Diff-Quik-stained cytospins prepared from
each lavage. Data were analyzed from two different ex-
periments. The results for total counts and differentials
were similar in all experiments and were pooled. The
graphs depict averages for each group ⫾ SEM.

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


form nitrate (NO3⫺). To measure total in vivo NO activity in samples, tration. Twenty micrograms of each extract was analyzed according to the
NO3⫺ was first converted to NO2⫺, and then total NO2⫺ concentration was manufacturer’s instructions for the ELISA kit. The absorbance at 450 nm
measured. Peritoneal lavage fluid from naive and CLP mice was mixed was read, and background absorbance (negative control) was subtracted
with an equal volume of nitrate reductase mixture (1 mg/ml NADPH and from each sample.
0.4 U/ml nitrate reductase (Sigma-Aldrich) in 0.2 M KH2PO4, pH ⫽ 7.4)
and incubated overnight at 37°C/5%CO2. The next day, 40 ␮l of 0.5% Statistics
sulfanilamide and 0.05% napthylethylenediamine dihydrochloride in 2.5%
In the case of survival studies, the log rank test was used to evaluate
H3PO4 were added to each standard and sample, and the absorbance was
significance. All other data are shown as the mean ⫾ SEM. Bacterial CFU
read at 550 nm. Total NO2⫺ in peritoneal lavage fluid was measured in
data were analyzed with the nonparametric Mann-Whitney U test, while all
three different experiments. Each experiment exhibited similar results and
other data were subjected to the unpaired Student’s t test. A p value ⬍0.05
was consequently pooled.
(ⴱ) was considered statistically significant. Highly significant p values
In vitro LPS stimulation ⬍0.005 (ⴱⴱ) and ⬍0.0005 (ⴱⴱⴱ) were also indicated.

Naive peritoneal macrophages were plated at 5 ⫻ 105/well of a 24-well cell Results


culture plate (Corning Glass). After resting overnight, macrophages were
CCR1-deficient mice were significantly less susceptible to
treated with medium alone or 1 ␮g/ml LPS O55:B5 (Sigma-Aldrich). Cells
were incubated 24 h at 37°C/5% CO2, after which cell-free supernatants CLP-induced lethality
were collected. Cytokine/chemokine production was measured by ELISA To determine the role of CCR1 in a murine model of experimental
(as described above) in at least three experiments. All experiments had
similar results. sepsis, both WT BALB/c (CCR1⫹/⫹) and CCR1⫺/⫺ mice were
subjected to CLP. In female WT mice, 21-gauge CLP was almost
NF-␬B p65 ELISA 100% lethal (19 of 20 mice) as early as 5 days after CLP (Fig. 1A).
Four WT and CCR1⫺/⫺ mice were injected with 3 ml of sterile 4% thio- However, female CCR1⫺/⫺ mice were significantly protected
glycolate i.p. After 5 days, peritoneal cells were isolated, as described against CLP-induced lethality. They experienced only 41% mor-
above, and 107 macrophages were plated per 100-mm cell culture plate tality (7 of 17 mice) at day 5 and 59% mortality (10 of 17 mice)
(Corning Glass). Macrophages were rested overnight and then treated with at day 7, demonstrating 54 and 36% increased survival, respec-
10 –100 ng/ml CCL5, 1 ␮g/ml LPS, or both CCL5 and LPS. Untreated
cells were included as a control. Four hours later, nuclear extracts were tively. Due to enhanced susceptibility of male mice to CLP, a
prepared according to manufacturer’s instructions and analyzed using the 26-gauge needle was used to induce sepsis in these mice. As
TransAM NF-␬B p65 transcription factor assay kit (Active Motif, Carls- shown in Fig. 1B, even in this less severe form of sepsis, complete
bad, CA). Briefly, cells were washed and scraped into ice-cold PBS con- mortality was observed in WT mice at the end of the experiment.
taining 6.25 mM NaF, 12.5 mM ␤-glycerophosphate, 12.5 mM para-nitro-
Once again, male CCR1⫺/⫺ mice were more resistant to these
phenyl phosphate, and 1.25 mM NaV03 (Sigma-Aldrich). Cells were
pelleted and resuspended in ice-cold hypotonic buffer (20 mM HEPES, pH lethal effects. None of the CCR1⫺/⫺ mice died until 3 days post-
7.5, 5 mM NaF, 10 ␮M Na2MoO4, and 1 mM EDTA; Sigma-Aldrich). CLP, at which time 90% of the WT mice (9 of 10 mice) were dead.
After 15 min on ice, cells were lysed with 0.5% Nonidet P-40 (Sigma- At day 7 post-CLP, 40% of the CCR1⫺/⫺ mice survived (4 of 10
Aldrich). The nuclear fraction was pelleted at 15,000 ⫻ g for 15 min and mice) compared with 0% of the WT mice. The rest of this inves-
resuspended in complete lysis buffer. After 30 min of agitation at 4°C,
debris was removed by pelleting, and nuclear extracts were snap frozen in tigation focused on identifying the immunomodulatory mechanism
a dry ice/ethanol bath. Extracts were stored at ⫺80°C. Before the ELISA, in CCR1⫺/⫺ mice that resulted in decreased CLP susceptibility
extracts were subjected to Bradford analysis to determine protein concen- and increased survival.
6942 CCR1 IN SEPSIS

FIGURE 4. Cytokine and che-

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


mokine expression were signifi-
cantly altered in CCR1⫺/⫺ mice
following CLP. At 0, 4, 8, or 24 h
post-CLP, mice were sacrificed
and bled, and 2-ml peritoneal la-
vages were performed. Concen-
trations of cytokines/chemokines
in serum (A) and lavage fluid (B)
from WT BALB/c mice (f) and
CCR1⫺/⫺ mice (䡺) were mea-
sured using specific sandwich
ELISAs. The data from three in-
dependent experiments were sim-
ilar, and representative data are
shown (n ⫽ 3–5 per group). ⴱ,
p ⬍ 0.05; ⴱⴱ, p ⬍ 0.005; ⴱⴱⴱ,
p ⬍ 0.0005, compared with sim-
ilarly treated WT mice.

CCR1⫺/⫺ mice displayed increased bacterial clearance mice was significantly lower than WT mice (Fig. 2A). At 8 h
subsequent to accelerated production of cytotoxic mediators post-CLP, CCR1⫺/⫺ mice had an average of 0.11 ⫻ 102 CFU/10
following CLP ␮l peritoneal lavage fluid, while WT levels were significantly el-
The process of CLP causes leakage of the polymicrobial flora into evated (2.14 ⫻ 103 CFU/10 ␮l). At 24 h post-CLP, mean bacterial
the peritoneal cavity. Peritoneal lavages were performed on mice burdens were 28-fold higher in WT mice (2.66 ⫻ 106 CFU/10 ␮l)
to assess the degree of local infection over time, while blood was compared with CCR1⫺/⫺ mice (9.52 ⫻ 104 CFU/10 ␮l). In WT
collected to measure systemic spread of the infection. The amount mice, decreased clearance and containment of bacteria at the local
of bacteria recovered from the peritoneal cavities of CCR1⫺/⫺ site of infection (peritoneal cavity) contributed to exacerbated
The Journal of Immunology 6943

spread throughout the animal (Fig. 2B). WT mice had dramatically CCR1⫺/⫺ macrophages had enhanced innate immune responses
higher systemic levels of bacteria at both 8 and 24 h post-CLP Although there were no apparent differences in leukocyte recruit-
(1.25 ⫻ 103 and 5.25 ⫻ 103 CFU/10 ␮l, respectively), when com- ment to the peritoneal cavity, key differences were observed in
pared with CCR1⫺/⫺ mice (0.10 ⫻ 102 and 2.80 ⫻ 102 CFU/10 bacterial clearance and cytokine/chemokine expression in
␮l, respectively). CCR1⫺/⫺ mice. These data implied that intrinsic differences ex-
Both macrophages and neutrophils produce a variety of nitrogen isted in the cellular activation of cells within the peritoneal cavity
and oxygen radicals that contribute to their repertoire of antimi- of these animals. The next series of experiments were designed to
crobial activity. Peritoneal lavage fluid was collected from mice at assess the specific effects of CCR1 deficiency on resident perito-
various times after CLP and evaluated for nitrite, a stable byprod- neal macrophages.
uct of NO (Fig. 2C). After CLP, peritoneal nitrite concentrations Peritoneal macrophages were isolated from naive WT and
steadily increased in WT mice and were highest at 24 h. In con- CCR1⫺/⫺ mice and infected in vitro with E. coli. Phagocytosis
trast, nitrite concentrations in CCR1⫺/⫺ mice peaked and were was 7.3-fold higher in CCR1⫺/⫺ macrophages (mean ⫽ 3.7 ⫻ 104
higher than in WT mice at 4 h after CLP (12.49 ⫾ 2.80 vs 6.24 ⫾ CFU/well) than WT macrophages (mean ⫽ 5.1 ⫻ 103 CFU/well)
0.66 ␮M in CCR1⫺/⫺ and WT mice, respectively). Nitrite con- after 1 h of infection (Fig. 5A). The combined stimulus of IFN-␥
centrations in CCR1⫺/⫺ mice remained steady without further in- and LPS was required to stimulate production and release of nitrite
crease over the 24-h period. At 24 h post-CLP, these levels were from WT macrophages (15.34 ⫾ 0.63 ␮M) in culture (Fig. 5B).
significantly lower than the amount of nitrite produced in WT mice However, CCR1⫺/⫺ macrophages were activated by IFN-␥ alone
(11.90 ⫾ 2.84 vs 44.51 ⫾ 11.38 ␮M in WT), directly correlating (14.86 ⫾ 0.56 ␮M) and demonstrated a greatly amplified response
with the decreased bacterial load, and, therefore, decreased stim- to the combination of IFN-␥ and LPS (36.9 ⫾ 1.1 ␮M).
ulus, present in CCR1⫺/⫺ mice at that time.

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


In other experiments, naive peritoneal macrophages were stim-
ulated in vitro with LPS. Supernatants were collected 24 h later
CCR1 was nonessential to the chemotaxis of leukocytes to the and assayed by ELISA. CCR1⫺/⫺ macrophages expressed signif-
peritoneal cavity in response to sepsis icantly higher constitutive levels of MIP-2, CCL2, CCL5, CCL6,
and CCL22 (Fig. 6). Although LPS stimulated substantial produc-
One obvious explanation for the increased bacterial clearance and tion of multiple cytokines and chemokines from WT macrophages,
accelerated nitrite production observed in CCR1⫺/⫺ mice was po- it provoked significantly higher levels from CCR1⫺/⫺ macro-
tential differences in the leukocyte populations recruited to the phages (Fig. 6).
peritoneal cavity following CLP. Therefore, peritoneal lavages
were examined from WT and CCR1⫺/⫺ mice at various times after
CLP to assess leukocyte infiltration (Fig. 3). In these studies, CLP
was a strong stimulus for recruitment of cells into the peritoneal
cavity; however, no differences were observed in total numbers of
leukocytes between the two groups (Fig. 3A). As expected, a ro-
bust neutrophilic response occurred quickly after CLP in WT mice
(peaking at 8 –24 h; Fig. 3B). No discrepancies were noted in
mononuclear cell and neutrophil recruitment between WT and
CCR1⫺/⫺ mice after CLP, indicating that enhanced bacterial clear-
ance in the CCR1⫺/⫺ mice was not due to changes in cellular
recruitment.

Temporal changes in expression and an altered


cytokine/chemokine profile were observed in CCR1⫺/⫺ mice
following CLP
Studies have shown that following CLP, the local and systemic
expression of many cytokines and chemokines is augmented.
Therefore, the cytokine profiles of naive and CLP-treated WT and
CCR1⫺/⫺ mice were measured in blood and peritoneal lavage fluid
at various times after surgery. CCR1⫺/⫺ mice produced signifi-
cantly higher levels of systemic IFN-␥ and CCL2 than WT mice
4 h after CLP (Fig. 4A). Lower levels of these cytokines, as well FIGURE 5. Macrophages deficient in CCR1 displayed enhanced phago-
as MIP-2 and KC, were observed later after CLP, compared with cytic and cytotoxic activity. A, Peritoneal cells were isolated from naive
WT. In general, peritoneal cytokine and chemokine expression WT (f) and CCR1⫺/⫺ mice (䡺) and plated at 106 macrophages per well
was either unchanged or decreased at later times (8 and 24 h) in (24-well plate). Nonadherent cells were removed, and macrophages were
CCR1⫺/⫺ mice when compared with WT mice (Fig. 4B). In WT infected with 106 E. coli for 1 h at 37°C. Extracellular bacteria were re-
mice, peritoneal TNF-␣ levels were induced at 4 h post-CLP and moved, cells were lysed, and concentrations of CFU were determined.
were significantly amplified throughout the 24-h period after CLP. Results were similar from two independent studies, and representative data
are shown. Horizontal bars represent the mean for each group (n ⫽ 5– 6).
Conversely, TNF-␣ peaked at 4 h in CCR1⫺/⫺ mice and was sig-
B, Naive macrophages were plated (5 ⫻ 105/well of 96-well plate) and
nificantly reduced in comparison with WT levels at 8 and 24 h
rested overnight. WT (f) and CCR1⫺/⫺ (䡺) macrophages were untreated
post-CLP. Several inflammatory chemokines (MIP-2, KC, CCL2, or stimulated with 100 ng/ml LPS, 20 U/ml IFN-␥, or LPS ⫹ IFN-␥ for
and CXCL9) were significantly higher in WT mice, compared with 48 h. Nitrite (NO2⫺) concentrations were measured in the supernatants
CCR1⫺/⫺ mice at 8 and/or 24 h post-CLP. Conversely, peritoneal (n ⱖ 3 per group). Similar results were obtained from three independent
CXCL10, CCL6, and CCL17 expression in CCR1⫺/⫺ mice was studies, and representative data are shown. ⴱⴱⴱ, p ⬍ 0.0005 compared with
significantly increased over WT expression at 8 h post-CLP. similarly treated WT macrophages.
6944 CCR1 IN SEPSIS

FIGURE 6. Cytokine/chemokine production was


significantly increased in CCR1⫺/⫺ macrophages. Na-
ive macrophages from WT (f) and CCR1⫺/⫺ mice (䡺)
were treated with medium or 1 ␮g/ml LPS for 24 h.
Supernatants were collected and clarified, and cytokine/
chemokine levels were measured by ELISA (n ⫽ 3 per

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


group). Results are presented as average for the
group ⫾ SEM. Representative data are shown for four
independent experiments. ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ 0.005;
ⴱⴱⴱ, p ⬍ 0.0005 compared with similarly treated WT
macrophages.

CCL5 increased lethality in CLP-induced sepsis CCL5 acted through CCR1 to increase NF-␬B activation and
CCR1 ⫺/⫺
mice were clearly protected against the lethal effects of CLP-induced inflammatory cytokine production
sepsis, and their macrophages demonstrated significantly enhanced When thioglycolate-elicited peritoneal macrophages were treated
innate immune responses. Because CCL3 and CCL6 were previ- with CCL5 in vitro, no significant differences were observed in the
ously recognized to play protective roles in this model of sepsis (7, NF-␬B activation of WT or CCR1⫺/⫺ cells; however, CCR1⫺/⫺
21), we investigated the role of CCL5 in this model. CCL5 ex- macrophages had slightly higher constitutive levels of activation
pression was strongly up-regulated following CLP in WT and (Fig. 9A). LPS induced similar levels of NF-␬B activation in WT
CCR1⫺/⫺ mice (Fig. 7). It was detected as early as 4 h in the and CCR1⫺/⫺ macrophages. In the context of LPS stimulation,
peritoneal cavity; however, peak expression was not observed until CCL5 caused a dose-dependent increase in NF-␬B activation in
at least 24 h after CLP in the peritoneal cavity and the serum of WT, but not CCR1⫺/⫺, macrophages. These data provide evidence
both mouse strains (Fig. 7). WT-CLP mice expressed significantly that CCL5 interacts primarily with CCR1 in vitro.
higher systemic levels of CCL5 at 24 h post-CLP than CCR1⫺/⫺ When rCCL5 was administered 2 h after CLP, peritoneal ex-
mice. When rCCL5 was administered 2 h post-CLP, day 4 mor- pression of IFN-␥ and MIP-2 was amplified 264 and 125%, re-
tality was significantly higher (88%) than in CLP-saline-treated spectively, in WT mice (Fig. 9B), while CCR1⫺/⫺ mice were un-
mice (59% mortality; p ⫽ 0.03; Fig. 8A). In contrast, when en- responsive to the effects of this ligand. WT production of
dogenous CCL5 was blocked with a specific goat anti-murine peritoneal KC, CCL22, and CXCL9 (53, 51, and 72%, respec-
CCL5 IgG 2 h prior and 48 h after CLP, survival was significantly tively) as well as systemic IL-12, IFN-␥, and KC (81, 69, and
improved (Fig. 8B). Two days after CLP, only 30% of control 104%, respectively) was also augmented to a lesser degree by
IgG-treated mice were alive, whereas 80% of mice treated with CCL5 treatment after CLP (data not shown). No changes were
anti-CCL5 IgG were remaining ( p ⫽ 0.0036). When CCR1⫺/⫺ observed in CCL3 expression following CCL5 administration in
mice were treated with rCCL5 2 h following CLP (Fig. 8C), no naive or CLP-treated WT or CCR1⫺/⫺ mice (data not shown).
significant effect was seen on the 4-day mortality (1 of 9 mice) Cytokine/Chemokine expression in CCR1⫺/⫺-CLP mice remained
when compared with saline-treated CCR1⫺/⫺-CLP mice (3 of 9 unchanged. In contrast, CCL5 appeared to have no effect on the
mice; p ⫽ 0.32). expression of protective chemokines, such as CXCL10 or CCL6,
The Journal of Immunology 6945

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


FIGURE 7. CLP induced local and systemic expression of CCL5 in WT
and CCR1⫺/⫺ mice. At 0, 4, 8, or 24 h post-CLP, mice were sacrificed and
bled, and 2-ml peritoneal lavages were performed. CCL5 was measured in
peritoneal wash (PW) fluid (A) and serum (B) from WT (f) and CCR1⫺/⫺
mice (䡺) using a specific ELISA. The data are representative of two in-
dependent studies (n ⫽ 3–5 per group). ⴱ, p ⬍ 0.05 compared with sim-
ilarly treated WT mice.

which continued to be expressed at significantly higher levels in


CCR1⫺/⫺ mice. These data independently verified the interaction
of CCL5 with CCR1 in a relevant sepsis model. Most likely, the
exaggerated inflammatory cytokine expression observed after CLP
in WT CCL5-treated mice was subsequent to the previously ob-
FIGURE 8. CCL5 significantly increased mortality of mice following
served augmentation of CCR1-dependent NF-␬B activation. CLP. A, Female WT BALB/c mice received saline (f) or 1 ␮g of CCL5
(䡺) i.p. 2 h post-CLP. B, WT mice received 0.5 mg of control (f) or
Discussion anti-murine CCL5 IgG (䡺) i.p. 2 h before and 48 h after CLP. Similar
Mounting evidence emphasizes the importance of both activated results were observed in replicate studies; therefore, data were pooled. C,
neutrophils and macrophages for an effective host response against Female CCR1⫺/⫺ mice received saline (f) or 1 ␮g of CCL5 (䡺) i.p. 2 h
pathogen infections, including sepsis (8). CCR1, a widely ex- post-CLP. Survival was followed for 4 days after surgery (n ⫽ 5–10 per
pressed chemokine receptor found on both of these cell types, has group).
proven to be fundamental in cellular trafficking and activation (13)
as well as performing a central role in modulating the cytokine
balance in several immunological models (16). In light of the shown to exert pro- and anti-inflammatory effects during the septic
widespread distribution of this receptor, especially on phagocytic response (7, 8, 28).
cells, we were interested in the role that CCR1 plays in the host IFN-␥ is a powerful inducer of NO, and, therefore, promotes
response to sepsis. This study clearly indicated that CCR1, while bacterial clearance and host survival (29). CCR1-deficient mice
dispensable for leukocyte recruitment, was a pivotal focus of the responded more rapidly to CLP and attained earlier peak systemic
immune response to sepsis in which the balance of pro- and anti- expression of IFN-␥ (4 vs 24 h in WT). Simultaneously, increased
inflammatory cytokines rested. Additionally, CCR1 regulated concentrations of nitrite were measured in the peritoneal cavities
macrophage responses to bacterial infection and LPS stimulation. of CCR1⫺/⫺ CLP mice. Consequently, significantly lower local
CCL5 functioned in a CCR1-dependent manner to detrimentally and systemic levels of bacteria were found in these mice, com-
amplify proinflammatory cytokine production and increase sepsis- pared with WT-CLP mice, indicating that an accelerated antimi-
related mortality. crobial response was highly effective. Despite radically amplified
Cytokines are often described as double-edged swords in that they IFN-␥ and nitrite production at later times after CLP (8 and 24 h),
are required for recruitment and activation of cells necessary for re- WT mice were not able to effectively control the infection, once
sponding to a foreign insult; however, when produced in excess pre- again emphasizing the importance of an early response.
cipitate damage against the host for which they were produced to CLP also induced quicker systemic expression of CCL2 in
protect. A successful response to infection, as in sepsis, is comprised CCR1⫺/⫺ mice (4 h), compared with 24-h peak expression in
of a complex balance of pro- and anti-inflammatory cytokines. A local WT-CLP mice. CCL2 has previously been shown to increase CLP
type 1 cytokine-driven response (dominated by IFN-␥ and IL-12) is survival by promoting activation of macrophages and production
crucial to contain and eliminate the source of the infection (23), while of anti-inflammatory cytokines, protecting the host from excessive
a systemic type 2 cytokine response (i.e., IL-10) is essential to mod- inflammation and resulting tissue damage (28, 30). This immuno-
ulate the inflammatory response, protecting the host from tissue and modulation was observed in the CCR1⫺/⫺ mice at 8 and 24 h
organ injury (27). Several CC and CXC chemokines have also been post-CLP when systemic expression of inflammatory cytokines/
6946 CCR1 IN SEPSIS

FIGURE 9. CCL5 increased NF-␬B activation by


LPS in vitro and CLP-induced inflammatory cytokine
expression in vivo. A, Thioglycolate-elicited macro-
phages from WT (f) and CCR1⫺/⫺ mice (䡺) were
treated in culture with 0, 10, or 100 ng/ml CCL5 ⫾ LPS
for 4 h. Nuclear extracts were prepared and assayed by
ELISA for the p65 subunit of NF-␬B. Absorbance was
read at 450 nm. B, WT (f) and CCR1⫺/⫺ mice (䡺)
were subjected to 21-gauge CLP, followed 2 h later by
i.p. injection with saline or 1 ␮g of CCL5. Additional

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


mice received saline or CCL5 with no surgery. Four
hours after administration of saline/CCL5 (6 h post-
CLP), mice were sacrificed and peritoneal lavages were
performed. Cytokine/chemokine concentrations were
measured in lavage fluid using specific ELISAs. ⴱ, p ⬍
0.05; ⴱⴱ, p ⬍ 0.005; ⴱⴱⴱ, p ⬍ 0.0005 compared with
similarly treated WT mice (n ⫽ 5 per group); #, p ⬍
0.05 compared with CLP (without CCL5).

chemokines and nitrite production were significantly lower than in 8, 11). In the absence of altered leukocyte recruitment after CLP,
WT-CLP mice. Similar regulation occurred in the peritoneal cav- changes in bacterial clearance and cytokine expression suggested
ity, where inflammatory mediators peaked at 4 h post-CLP in intrinsic differences in the activation of WT and CCR1⫺/⫺ peri-
CCR1⫺/⫺ mice, but continued to rise throughout the 24-h period toneal cells. In vitro analyses of CCR1⫺/⫺ macrophages demon-
after CLP in WT mice. TNF-␣, while critical to the immune re- strated overall enhanced innate responses, including increased
sponse, is able to solely initiate many of the immunopathological phagocytosis, cytotoxicity, and cytokine/chemokine production.
features of septic shock (31), and, therefore, must be rapidly reg- These data support the accelerated responses seen in CCR1⫺/⫺
ulated. Its continued expression may serve to propagate the ex- mice after CLP and emphasize the central function that peritoneal
tended inflammatory response observed after CLP in WT mice. macrophages execute in the innate response following sepsis.
Interestingly, CCR1⫺/⫺ mice showed significantly increased In this study, we confirmed that the CCR1 ligands, CCL3 and
peritoneal expression of CXCL10, CCL6, and CCL17 when com- CCL6, were produced in WT mice following CLP (7, 20). How-
pared with WT mice at 8 –24 h post-CLP. Studies from our labo- ever, this study was the first to explore the expression and role of
ratory previously demonstrated major protective roles for both the CCR1 ligand, namely CCL5, in a CLP model of sepsis. Pre-
CXCL10 and CCL6 against CLP-induced lethality (7, 11). viously, it was shown that CCL5 was elevated in septic patients as
CXCL10 acted through an unidentified mechanism, while CCL6 well as volunteers treated with LPS (19, 33). In a colon ascendens
augmented peritoneal macrophage activity and reduced bacterial stent peritonitis model of sepsis, CCL5 induction was present dur-
leak from the gut. The role of CCL17, a predominantly recognized ing renal failure (34). In the CLP model, local and systemic ex-
Th2 lymphocyte chemoattractant, in sepsis is unknown. Its expres- pression of CCL5 was strongly provoked in WT mice. CCR1⫺/⫺
sion may contribute to the regulation of the proinflammatory type expression of CCL3 and CCL5 was equal to or less than WT
1 response in CCR1⫺/⫺ mice after CLP. expression at all times after CLP. In contrast, CCL6 expression
Peritoneal macrophages are the principal resident cells, and are was much higher in CCR1⫺/⫺ than WT mice at 8 and 24 h after
known to play an essential role in infection and inflammation by CLP, supporting the idea that CCL6 has only one receptor in vivo
producing a large variety of mediators, including cytokines/che- (18). The elevations in CCL6 presumably reflected a continued
mokines, in response to infectious bacteria or bacterial components accumulation of ligand in the absence of functional receptor to
such as LPS (32). Previous work from our laboratory has illus- bind and regulate its production. Both CCL3 and CCL5 are known
trated the importance of these cells in the CLP model of sepsis (7, to interact with multiple receptors, principally CCR1 and CCR5
The Journal of Immunology 6947

(12). In the absence of CCR1, mice were protected against the to CCL3 and CCL6, other previously identified CCR1 ligands.
deleterious effects of sepsis, indicating that CCR1 is a central Exogenous CCL5 administered after CLP induced a hyperinflam-
player in this injurious response. Its ligand counterpart was pre- matory response dominated by IFN-␥ in WT, but not in CCR1⫺/⫺
dicted to have a similar detrimental effect. Considering the previ- mice, indicating that the deleterious actions of CCL5 were CCR1
ously established protective roles of CCL3 (21) and CCL6 (7) in dependent. Thus, these data suggested that CCR1 and CCL5 are
this model, CCL5 was the obvious ligand to consider. In our both key mediators in modulating the innate inflammatory re-
model, CCL5 demonstrated a negative effect on CLP-associated sponse generated during a septic insult and are potential targets for
survival in wild-type, but not CCR1⫺/⫺ mice, implicating its in- immunotherapy in septic patients.
volvement as another key mediator of the deleterious host
response. References
CCR1⫺/⫺ mice and macrophages were nonresponsive to CCL5, 1. Angus, D. C., W. T. Linde-Zwirble, J. Lidicker, G. Clermont, J. Carcillo, and
indicating that CCL5 interacted directly with CCR1. Although M. R. Pinsky. 2001. Epidemiology of severe sepsis in the United States: analysis
CCL3 is generally thought to be the principal in vivo ligand of incidence, outcome, and associated costs of care. Crit. Care Med. 29:1303.
2. Zanotti, S., and A. Kumar. 2002. Cytokine modulation in sepsis and septic shock.
for CCR1, other studies have demonstrated CCL5 action through Exp. Opin. Investig. Drugs 11:1061.
CCR1. CCL5 was identified as a potent agonist that stimulated 3. Esmon, C. T. 2002. Protein C pathway in sepsis. Ann. Med. 34:598.
CCR1-dependent chemotaxis of mast cells (35). CCR1 was also 4. Bhatia, M., and S. Moochhala. 2004. Role of inflammatory mediators in the
pathophysiology of acute respiratory distress syndrome. J. Pathol. 202:145.
recognized as the major receptor for CCL5 binding in memory and 5. Makhija, R., and A. N. Kingsnorth. 2002. Cytokine storm in acute pancreatitis.
naive CD4⫹ T cells, as well as acting in CCL5-induced chemo- J. Hepatobiliary Pancreat. Surg. 9:401.
taxis of memory T cells (36). Although CCL5 has been predom- 6. Baggiolini, M., B. Dewald, and B. Moser. 1994. Interleukin-8 and related che-
motactic cytokines: CXC and CC chemokines. Adv. Immunol. 55:97.

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


inantly studied as an important mediator of type 2-driven diseases, 7. Steinhauser, M. L., C. M. Hogaboam, A. Matsukawa, N. W. Lukacs,
it has become apparent that the regulatory function of CCL5 has R. M. Strieter, and S. L. Kunkel. 2000. Chemokine C10 promotes disease reso-
broader implications than previously recognized. CCL5 is a potent lution and survival in an experimental model of bacterial sepsis. Infect. Immun.
68:6108.
stimulus for macrophage recruitment into the lung following en- 8. Matsukawa, A., C. M. Hogaboam, N. W. Lukacs, P. M. Lincoln, H. L. Evanoff,
dotoxemia (37) as well as into synovial tissue during rheumatoid and S. L. Kunkel. 2000. Pivotal role of the CC chemokine, macrophage-derived
arthritis, propagating the chronic inflammation that is characteris- chemokine, in the innate immune response. J. Immunol. 164:5362.
9. Cummings, C. J., T. R. Martin, C. W. Frevert, J. M. Quan, V. A. Wong,
tic of the disease (38). Intradermal injection of rCCL5 results in S. M. Mongovin, T. R. Hagen, K. P. Steinberg, and R. B. Goodman. 1999.
pathology characterized by monocytic and eosinophilic inflamma- Expression and function of the chemokine receptors CXCR1 and CXCR2 in
sepsis. J. Immunol. 162:2341.
tory responses (39). CCL5 acts not only as a chemoattractant, but
10. Chvatchko, Y., A. J. Hoogewerf, A. Meyer, S. Alouani, P. Juillard, R. Buser,
operates synergistically with IFN-␥ to activate macrophages, NK F. Conquet, A. E. Proudfoot, T. N. Wells, and C. A. Power. 2000. A key role for
cells, and T cells, alluding to its central role in modulating type 1 CC chemokine receptor 4 in lipopolysaccharide-induced endotoxic shock. J. Exp.
Med. 191:1755.
immune responses (40). 11. Ness, T. L., C. M. Hogaboam, R. M. Strieter, and S. L. Kunkel. 2003. Immu-
In the sepsis model, CCL5 acted to amplify the expression of nomodulatory role of CXCR2 during experimental septic peritonitis. J. Immunol.
inflammatory cytokines in WT mice without affecting the produc- 171:3775.
12. Matsukawa, A., N. W. Lukacs, C. M. Hogaboam, S. W. Chensue, and
tion of protective mediators such as CXCL10 and CCL6. CCL5 S. L. Kunkel. 2001. III. Chemokines and other mediators. 8. Chemokines and
triggered an overwhelming IFN-␥ response within the peritoneal their receptors in cell-mediated immune responses in the lung. Microsc. Res.
cavity after CLP in WT mice. Although the protective function of Tech. 53:298.
13. Shang, X., B. Qiu, K. A. Frait, J. S. Hu, J. Sonstein, J. L. Curtis, B. Lu, C. Gerard,
IFN-␥ has been demonstrated in a number of infection models (41) and S. W. Chensue. 2000. Chemokine receptor 1 knockout abrogates natural
as well as in the CCR1⫺/⫺ mice following CLP, overproduction or killer cell recruitment and impairs type-1 cytokines in lymphoid tissue during
abundance of this cytokine directly after CLP significantly in- pulmonary granuloma formation. Am. J. Pathol. 157:2055.
14. Rodriguez-Sosa, M., L. E. Rosas, L. I. Terrazas, B. Lu, C. Gerard, and
creases lethality (42, 43). In our model, CCL5 also up-regulated A. R. Satoskar. 2003. CC chemokine receptor 1 enhances susceptibility to Leish-
MIP-2 production, as well as marginally affecting the expression mania major during early phase of infection. Immunol. Cell Biol. 81:114.
of other inflammatory cytokines/chemokines. It is well-established 15. Domachowske, J. B., C. A. Bonville, J. L. Gao, P. M. Murphy, A. J. Easton, and
H. F. Rosenberg. 2000. The chemokine macrophage-inflammatory protein-1␣
that MIP-2 contributes to neutrophil recruitment, resulting in host and its receptor CCR1 control pulmonary inflammation and antiviral host defense
injury and CLP mortality (20). CCL5 may augment type 1 cyto- in paramyxovirus infection. J. Immunol. 165:2677.
kine production directly or indirectly by decreasing anti- 16. Gao, J. L., T. A. Wynn, Y. Chang, E. J. Lee, H. E. Broxmeyer, S. Cooper,
H. L. Tiffany, H. Westphal, J. Kwon-Chung, and P. M. Murphy. 1997. Impaired
inflammatory type 2 cytokine expression. Given that the levels of host defense, hematopoiesis, granulomatous inflammation and type 1-type 2 cy-
type 2 cytokines are very low during the acute phase after CLP, it tokine balance in mice lacking CC chemokine receptor 1. J. Exp. Med. 185:1959.
17. Lukacs, N. W., S. H. Oliveira, and C. M. Hogaboam. 1999. Chemokines and
is more likely that CCL5 acts directly on the expression of type 1 asthma: redundancy of function or a coordinated effort? J. Clin. Invest. 104:995.
cytokines. Locati et al. (44) have previously shown that CCL5 18. Ma, B., Z. Zhu, R. J. Homer, C. Gerard, R. Strieter, and J. A. Elias. 2004. The
stimulation of human monocytes in vitro induces RNA expression C10/CCL6 chemokine and CCR1 play critical roles in the pathogenesis of IL-
13-induced inflammation and remodeling. J. Immunol. 172:1872.
of key proinflammatory mediators IL-1␤ and IL-8 (human homo- 19. Cavaillon, J. M., M. Adib-Conquy, C. Fitting, C. Adrie, and D. Payen. 2003.
logue of MIP-2) as well as other inflammatory chemokines such as Cytokine cascade in sepsis. Scand. J. Infect. Dis. 35:535.
CCL2 and CCL3. Additionally, in vitro LPS activation of the in- 20. Walley, K. R., N. W. Lukacs, T. J. Standiford, R. M. Strieter, and S. L. Kunkel.
1997. Elevated levels of macrophage inflammatory protein 2 in severe murine
flammatory p65 subunit of NF-␬B was amplified in a dose-depen- peritonitis increase neutrophil recruitment and mortality. Infect. Immun. 65:3847.
dent manner by CCL5, lending further credence to a direct effect 21. Takahashi, H., T. Tashiro, M. Miyazaki, M. Kobayashi, R. B. Pollard, and
of CCL5 on the inflammatory response. F. Suzuki. 2002. An essential role of macrophage inflammatory protein 1␣/CCL3
on the expression of host’s innate immunities against infectious complications.
In summary, we have shown that genetic deletion of CCR1 in- J. Leukocyte Biol. 72:1190.
creased survival in septic mice. Although no effect on leukocyte 22. Gerard, C., J. L. Frossard, M. Bhatia, A. Saluja, N. P. Gerard, B. Lu, and
recruitment was observed, CCR1⫺/⫺ mice exhibited an acceler- M. Steer. 1997. Targeted disruption of the ␤-chemokine receptor CCR1 protects
against pancreatitis-associated lung injury. J. Clin. Invest. 100:2022.
ated, effective antimicrobial response that was quickly down-reg- 23. Steinhauser, M. L., C. M. Hogaboam, N. W. Lukacs, R. M. Strieter, and
ulated following bacterial clearance. Peritoneal macrophages from S. L. Kunkel. 1999. Multiple roles for IL-12 in a model of acute septic peritonitis.
CCR1⫺/⫺ mice displayed enhanced innate immune responses that J. Immunol. 162:5437.
24. Belperio, J. A., M. D. Burdick, M. P. Keane, Y. Y. Xue, J. P. Lynch III,
were also differentially regulated. We demonstrated that CCL5 B. L. Daugherty, S. L. Kunkel, and R. M. Strieter. 2000. The role of the CC
production after CLP had a negative effect on survival, in contrast chemokine, RANTES, in acute lung allograft rejection. J. Immunol. 165:461.
6948 CCR1 IN SEPSIS

25. Evanoff, H. L., M. D. Burdick, S. A. Moore, S. L. Kunkel, and R. M. Strieter. 36. Sato, K., H. Kawasaki, C. Morimoto, N. Yamashima, and T. Matsuyama. 2002.
1992. A sensitive ELISA for the detection of human monocyte chemoattractant An abortive ligand-induced activation of CCR1-mediated downstream signaling
protein-1 (MCP-1). Immunol. Invest. 21:39. event and a deficiency of CCR5 expression are associated with the hyporespon-
26. Green, S. J., C. A. Nacy, R. D. Schreiber, D. L. Granger, R. M. Crawford, siveness of human naive CD4⫹ T cells to CCL3 and CCL5. J. Immunol.
M. S. Meltzer, and A. H. Fortier. 1993. Neutralization of ␥ interferon and tumor 168:6263.
necrosis factor ␣ blocks in vivo synthesis of nitrogen oxides from L-arginine and 37. VanOtteren, G. M., R. M. Strieter, S. L. Kunkel, R. Paine III, M. J. Greenberger,
protection against Francisella tularensis infection in Mycobacterium bovis BCG-
J. M. Danforth, M. D. Burdick, and T. J. Standiford. 1995. Compartmentalized
treated mice. Infect. Immun. 61:689.
expression of RANTES in a murine model of endotoxemia. J. Immunol.
27. Sewnath, M. E., D. P. Olszyna, R. Birjmohun, F. J. ten Kate, D. J. Gouma, and
154:1900.
T. van Der Poll. 2001. IL-10-deficient mice demonstrate multiple organ failure
and increased mortality during Escherichia coli peritonitis despite an accelerated 38. Shadidi, K. R., T. Aarvak, J. E. Henriksen, J. B. Natvig, and K. M. Thompson.
bacterial clearance. J. Immunol. 166:6323. 2003. The chemokines CCL5, CCL2 and CXCL12 play significant roles in the
28. Matsukawa, A., C. M. Hogaboam, N. W. Lukacs, P. M. Lincoln, R. M. Strieter, migration of Th1 cells into rheumatoid synovial tissue. Scand. J. Immunol.
and S. L. Kunkel. 1999. Endogenous monocyte chemoattractant protein-1 57:192.
(MCP-1) protects mice in a model of acute septic peritonitis: cross-talk between 39. Meurer, R., G. Van Riper, W. Feeney, P. Cunningham, D. Hora, Jr.,
MCP-1 and leukotriene B4. J. Immunol. 163:6148. M. S. Springer, D. E. MacIntyre, and H. Rosen. 1993. Formation of eosinophilic
29. Moreira, A. L., L. Tsenova, P. J. Murray, S. Freeman, A. Bergtold, L. Chiriboga, and monocytic intradermal inflammatory sites in the dog by injection of human
and G. Kaplan. 2000. Aerosol infection of mice with recombinant BCG secreting RANTES but not human monocyte chemoattractant protein 1, human macro-
murine IFN-␥ partially reconstitutes local protective immunity. Microb. Pathog. phage inflammatory protein 1␣, or human interleukin 8. J. Exp. Med. 178:1913.
29:175.
40. Dorner, B. G., A. Scheffold, M. S. Rolph, M. B. Huser, S. H. Kaufmann,
30. Matsukawa, A., C. M. Hogaboam, N. W. Lukacs, P. M. Lincoln, R. M. Strieter,
and S. L. Kunkel. 2000. Endogenous MCP-1 influences systemic cytokine bal- A. Radbruch, I. E. Flesch, and R. A. Kroczek. 2002. MIP-1␣, MIP-1␤, RANTES,
ance in a murine model of acute septic peritonitis. Exp. Mol. Pathol. 68:77. and ATAC/lymphotactin function together with IFN-␥ as type 1 cytokines. Proc.
31. Cohen, J. 2002. The immunopathogenesis of sepsis. Nature 420:885. Natl. Acad. Sci. USA 99:6181.
32. Suzuki, T., S. Hashimoto, N. Toyoda, S. Nagai, N. Yamazaki, H. Y. Dong, 41. Bohn, E., and I. B. Autenrieth. 1996. IL-12 is essential for resistance against
J. Sakai, T. Yamashita, T. Nukiwa, and K. Matsushima. 2000. Comprehensive Yersinia enterocolitica by triggering IFN-␥ production in NK cells and CD4⫹ T

Downloaded from http://www.jimmunol.org/ by guest on October 26, 2022


gene expression profile of LPS-stimulated human monocytes by SAGE. Blood cells. J. Immunol. 156:1458.
96:2584. 42. Echtenacher, B., M. A. Freudenberg, R. S. Jack, and D. N. Mannel. 2001. Dif-
33. Kopydlowski, K. M., C. A. Salkowski, M. J. Cody, N. van Rooijen, J. Major, ferences in innate defense mechanisms in endotoxemia and polymicrobial septic
T. A. Hamilton, and S. N. Vogel. 1999. Regulation of macrophage chemokine peritonitis. Infect. Immun. 69:7271.
expression by lipopolysaccharide in vitro and in vivo. J. Immunol. 163:1537.
34. Maier, S., K. Emmanuilidis, M. Entleutner, N. Zantl, M. Werner, K. Pfeffer, and 43. Miles, R. H., T. P. Paxton, D. J. Dries, and R. L. Gamelli. 1994. Interferon-␥
C. D. Heidecke. 2000. Massive chemokine transcription in acute renal failure due increases mortality following cecal ligation and puncture. J. Trauma 36:607.
to polymicrobial sepsis. Shock 14:187. 44. Locati, M., U. Deuschle, M. L. Massardi, F. O. Martinez, M. Sironi, S. Sozzani,
35. Juremalm, M., N. Olsson, and G. Nilsson. 2002. Selective CCL5/RANTES-in- T. Bartfai, and A. Mantovani. 2002. Analysis of the gene expression profile
duced mast cell migration through interactions with chemokine receptors CCR1 activated by the CC chemokine ligand 5/RANTES and by lipopolysaccharide in
and CCR4. Biochem. Biophys. Res. Commun. 297:480. human monocytes. J. Immunol. 168:3557.

You might also like