ArticlePDF Available

Murine Lung Cancer Increases CD4+ T Cell Apoptosis and Decreases Gut Proliferative Capacity in Sepsis

PLOS
PLOS ONE
Authors:

Abstract and Figures

Background Mortality is significantly higher in septic patients with cancer than in septic patients without a history of cancer. We have previously described a model of pancreatic cancer followed by sepsis from Pseudomonas aeruginosa pneumonia in which cancer septic mice have higher mortality than previously healthy septic mice, associated with increased gut epithelial apoptosis and decreased T cell apoptosis. The purpose of this study was to determine whether this represents a common host response by creating a new model in which both the type of cancer and the model of sepsis are altered. Methods C57Bl/6 mice received an injection of 250,000 cells of the lung cancer line LLC-1 into their right thigh and were followed three weeks for development of palpable tumors. Mice with cancer and mice without cancer were then subjected to cecal ligation and puncture and sacrificed 24 hours after the onset of sepsis or followed 7 days for survival. Results Cancer septic mice had a higher mortality than previously healthy septic mice (60% vs. 18%, p = 0.003). Cancer septic mice had decreased number and frequency of splenic CD4+ lymphocytes secondary to increased apoptosis without changes in splenic CD8+ numbers. Intestinal proliferation was also decreased in cancer septic mice. Cancer septic mice had a higher bacterial burden in the peritoneal cavity, but this was not associated with alterations in local cytokine, neutrophil or dendritic cell responses. Cancer septic mice had biochemical evidence of worsened renal function, but there was no histologic evidence of renal injury. Conclusions Animals with cancer have a significantly higher mortality than previously healthy animals following sepsis. The potential mechanisms associated with this elevated mortality differ significantly based upon the model of cancer and sepsis utilized. While lymphocyte apoptosis and intestinal integrity are both altered by the combination of cancer and sepsis, the patterns of these alterations vary greatly depending on the models used.
Content may be subject to copyright.
RESEARCH ARTICLE
Murine Lung Cancer Increases CD4+ T Cell
Apoptosis and Decreases Gut Proliferative
Capacity in Sepsis
John D. Lyons
1
, Rohit Mittal
1
, Katherine T. Fay
1
, Ching-Wen Chen
1
, Zhe Liang
1
, Lindsay
M. Margoles
2
, Eileen M. Burd
3
, Alton B. Farris
3
, Mandy L. Ford
4
, Craig M. Coopersmith
1
*
1Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA,
United States of America, 2Department of Internal Medicine and Emory Critical Care Center, Emory
University School of Medicine, Atlanta, GA, United States of America, 3Department of Pathology and
Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States of America,
4Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, GA,
United States of America
These authors contributed equally to this work.
*cmcoop3@emory.edu
Abstract
Background
Mortality is significantly higher in septic patients with cancer than in septic patients without a
history of cancer. We have previously described a model of pancreatic cancer followed by
sepsis from Pseudomonas aeruginosa pneumonia in which cancer septic mice have higher
mortality than previously healthy septic mice, associated with increased gut epithelial apo-
ptosis and decreased T cell apoptosis. The purpose of this study was to determine whether
this represents a common host response by creating a new model in which both the type of
cancer and the model of sepsis are altered.
Methods
C57Bl/6 mice received an injection of 250,000 cells of the lung cancer line LLC-1 into their
right thigh and were followed three weeks for development of palpable tumors. Mice with
cancer and mice without cancer were then subjected to cecal ligation and puncture and sac-
rificed 24 hours after the onset of sepsis or followed 7 days for survival.
Results
Cancer septic mice had a higher mortality than previously healthy septic mice (60% vs.
18%, p = 0.003). Cancer septic mice had decreased number and frequency of splenic
CD4+ lymphocytes secondary to increased apoptosis without changes in splenic CD8+
numbers. Intestinal proliferation was also decreased in cancer septic mice. Cancer septic
mice had a higher bacterial burden in the peritoneal cavity, but this was not associated with
alterations in local cytokine, neutrophil or dendritic cell responses. Cancer septic mice had
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 1/20
OPEN ACCESS
Citation: Lyons JD, Mittal R, Fay KT, Chen C-W,
Liang Z, Margoles LM, et al. (2016) Murine Lung
Cancer Increases CD4+ T Cell Apoptosis and
Decreases Gut Proliferative Capacity in Sepsis. PLoS
ONE 11(3): e0149069. doi:10.1371/journal.
pone.0149069
Editor: Philip Alexander Efron, University of Florida,
UNITED STATES
Received: August 26, 2015
Accepted: January 27, 2016
Published: March 28, 2016
Copyright: © 2016 Lyons et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper.
Funding: This work was supported by funding from
the National Institutes of Health (GM104323,
GM095442, GM072808, GM109779, GM113228).
The funders had no role in study design, data
collection and analysis, decision to publish, or
preparation of the manuscript.
Competing Interests: Dr. Coopersmith is the
president of the Society of Critical Care Medicine.
biochemical evidence of worsened renal function, but there was no histologic evidence of
renal injury.
Conclusions
Animals with cancer have a significantly higher mortality than previously healthy animals fol-
lowing sepsis. The potential mechanisms associated with this elevated mortality differ sig-
nificantly based upon the model of cancer and sepsis utilized. While lymphocyte apoptosis
and intestinal integrity are both altered by the combination of cancer and sepsis, the pat-
terns of these alterations vary greatly depending on the models used.
Introduction
Sepsis is the leading causes of death among critically ill patients in the United States with
between 230,000 and 370,000 people dying of the disease annually [1]. Patients with malig-
nancy are nearly ten times more likely to develop sepsis than the general population [2], and
cancer represents the most common co-morbidity in septic patients[35]. Sepsis is also the
leading cause of ICU admission in patients with cancer[6,7]. Importantly, cancer is also the co-
morbidity associated with the highest risk of death in sepsis, with hospital mortality exceeding
50% in patients with cancer and either severe sepsis or septic shock[5,79].
The etiology behind the increased mortality seen in cancer patients who develop sepsis com-
pared to previously healthy patients who develop sepsis is multifactorial[2,10]. While some
deaths are related to immunosuppression caused by cancer treatment such as chemotherapy or
radiation, others are likely related to a reduced ability of the host to appropriately respond to
infection in the setting of chronic systemic changes related to underlying malignancy. Animal
models of cancer, in isolation, demonstrate that not only is the tumor microenvironment
altered, but that systemic T cell exhaustion and generalized immune suppression are also
induced by cancer[11]. Further, the host response to a non-lethal infection is markedly altered
following cancer, with phenotypic exhaustion in T cells associated with increasing expression
of co-inhibitory receptors[12].
There are numerous similarities in the host response to both cancer and sepsis[13]. In an
attempt to understand why hosts with cancer have increased mortality following sepsis com-
pared to previously healthy hosts, we have described a model of pancreatic cancer followed by
sepsis from Pseudomonas aeruginosa pneumonia[14]. Mortality was higher in cancer septic
mice than previously healthy mice and this was associated with a decrease in T lymphocyte
apoptosis and an increase in both gut epithelial apoptosis and bacteremia. Interestingly, pre-
venting lymphocyte apoptosisa strategy associated with uniform success in other pre-clinical
models of sepsiswas associated with increased mortality in cancer septic mice[15].
Despite having a greater understanding of the pathophysiology of sepsis than ever before
[1618], there has been a remarkable inability to translate preclinical models of sepsis into
effective treatments at the bedside, where management is generally supportive yet non-selec-
tive, with the exception of targeted antimicrobial therapy[19]. One reason (of many) for the
failure of pre-clinical trials to translate into effective therapies for sepsis is that animal studies
are performed in a homogenous previously healthy population, whereas human studies are
performed on heterogeneous patients frequently with multiple co-morbidities. As such, we
sought to determine whether our previous pre-clinical findings in cancer and sepsis would be
generalizable if we altered both the type of cancer and the model of sepsis. To examine this, we
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 2/20
This does not alter the authors' adherence to PLOS
ONE policies on sharing data and materials.
developed a new clinically relevant model of lung cancer followed by sepsis induced by cecal
ligation and puncture.
Materials and Methods
Animals
Male and female C57BL/6 mice were used in all experiments, with gender matching between
experimental and control groups. Animals were 68 weeks of age prior to initiation of experi-
ments. A subset of animals were then injected with tumor cells (details below) and all mice
were then watched for an additional three weeks before a subset were subjected to cecal ligation
and puncture (CLP, also detailed below) at which time they were watched for 17 days depend-
ing on whether they were used for non-survival or survival experiments. Thus animals were a
minimum of 9 weeks old and a maximum of 12 weeks old at time of sacrifice. Experiments
were performed in accordance with the National Institutes of Health Guidelines for the Use of
Laboratory Animals and were approved by the Institutional Animal Care and Use Committee
at Emory University School of Medicine (Protocol DAR-2001875-082815BN). All animals
were housed in an approved university animal facility and were given free access to food and
water throughout. Animals that were injected with tumor cells were monitored to ensure that
tumors did not ulcerate and did not impede animal ambulation according to the Emory
IACUC guidelines for tumor burden. Following CLP, all animals received buprenorphine post-
operatively in an attempt to minimize animal suffering. For non-survival studies, animals were
sacrificed 24 hours post-operatively via asphyxiation by CO2 or exsanguination under deep
ketamine anesthesia. A different subset of animals was followed for survival for 7 days post-
operatively. During this survival experiment, animals were checked twice daily. In addition to
observing the same endpoints outlined above surrounding tumor growth, animals were also
checked to determine if they were moribund related to operation. Animals that either met
tumor endpoints or were moribund were sacrificed using humane endpoints. Moribund ani-
mals were identified as follows: a) surgical complications unresponsive to immediate interven-
tion (wound dehiscence, bleeding, infection), b) medical conditions unresponsive to treatment
such as self-mutilation, severe respiratory distress, icterus, major organ failure or intractable
diarrhea, or c) clinical or behavioral signs unresponsive to appropriate intervention persisting
for 1 day including significant inactivity, labored breathing, sunken eyes, hunched posture,
piloerection/matted fur, one or more unresolving skin ulcers, and abnormal vocalization when
handled. Animals that survived 7 days post-operatively were sacrificed at the conclusion of this
experiment using asphyxiation by CO2.
Cancer model
A murine lung carcinoma cell line (LLC1, American Type Culture Collection, Manassas, VA)
was cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum, 1% gluta-
mine, 1% penicillin-streptomycin and 1%4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid.
The rationale for using a lung cancer cell line is that lung cancer has the second highest mortal-
ity for solid tumors in septic patients (2) (pancreatic cancer is the highest and was used for our
prior experiments in sepsis and cancer). After expansion, cancer cells were dissociated from
growth flasks via incubation with 0.25% trypsin, washed, centrifuged for 10 minutes at 1500
RPM and then re-suspended in phosphate buffered solution (PBS) to a final concentration of
250,000 cells per 0.2 ml (live cells selected via trypan blue examination). Mice randomized to
receive cancer had a single subcutaneous injection of 250,000 tumor cells along the right inner
thigh (cancer group) and were followed for 3 weeks prior to CLP to allow for tumor growth.
Control mice were unmanipulated and thus had no intervention prior to CLP (previously
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 3/20
healthy group) but were watched for an identical time as cancer mice so animals would be age
matched.
Sepsis model and experimental groups
A subset of cancer mice and previously healthy mice were then subjected to CLP, an established
model of polymicrobial peritonitis[20]. Briefly, under isoflurane anesthesia, a small midline
abdominal incision was made, and the cecum was exteriorized and ligated below the ileocecal
valve, avoiding intestinal obstruction. The cecum was punctured twice with a 25 gauge needle
and squeezed gently to extrude a small amount of stool. After placing the cecum back in the
abdomen, the abdominal wall was closed in layers. Immediately following CLP, mice received
subcutaneous injections of a) fluids (1ml of 0.9% saline) to account for insensible losses, b)
antibiotics (50 mg/kg of ceftriaxone, Sigma-Aldrich, St. Louis, MO and 35 mg/kg metronida-
zole, Apotex Corp, Weston, FL) to mimic the clinical scenario where septic patients receive
antimicrobial therapy and c) pain medication (0.1 mg/kg buprenex, McKesson Medical, San
Francisco, CA) to minimize pain and suffering. Animals were either followed 7 days for sur-
vival or sacrificed at 24 hours for sample collection. Antibiotics were re-dosed at 12, 24 and 36
hours after surgery in survival studies.
We have previously published an extensive immunological assessment of lung cancer in
unmanipulated mice [11] so did not repeat those studies. However, we did not previously have
data on many of the outcomes assayed in this study and so performed experiments in both sep-
tic and non-septic animals where appropriate, in order to understand the impact of cancer in
isolation, sepsis in isolation, and the combination of cancer and sepsis. The following is the ter-
minology used for each experimental group: a) unmanipulated (mice that received neither can-
cer nor sepsis), b) cancer (mice that received tumor cell injection alone), c) previously healthy
septic (mice that underwent CLP alone without prior intervention), and d) cancer septic (mice
with tumor cell injection followed three weeks later by CLP).
Leukocyte analysis
Phenotypic flow cytometric analysis of leukocytes was performed on processed cellular suspen-
sions of splenocytes (whole spleens removed at time of sacrifice) or peritoneal fluid (2.5ml PBS
injected into peritoneum and withdrawn after 5 seconds of gentle agitation). The number of
cells per ml of suspension was calculated utilizing a Nexcelom Auto Cellometer, the results of
which were used to determine absolute cell numbers. The following antibodies were used to
stain cells prior to analysis: for peritoneal fluid, anti-GR1.1 FITC (BD Bioscience, San Jose,
CA), anti-Cd11b PerCP (BioLegend, San Diego, CA), anti-Cd11c PeCy7 (eBioscience, San
Diego, CA), and anti-MHC II APC Cy7 (eBioscience); for splenocyte phenotyping, anti-F4/80
PerCP (BioLegend), anti-Cd11c PeCy7 (eBioscience), anti-Cd11b APC (eBioscience), anti-
B220 Alexa700 (BD Bioscience), and anti-MHC II APC Cy7 (eBioscience).
To determine the frequency of apoptotic lymphocytes, splenocytes were collected from sac-
rificed animals and processed to a suspension of 1x10
7
cells/ml, and 1x10
6
splenocytes were
then processed using a commercially available Annexin V and 7-AAD kit (BioLegend) follow-
ing manufacturers instructions. Cells were then stained with anti-CD4-PO (Invitrogen, Carls-
bad, CA), anti-CD8-PB (eBioscience) to determine the frequency of pro-apoptotic CD4 and
CD8 T cells. A gating strategy excluded dead cells staining positive for 7-AAD from analysis.
For samples undergoing intracellular cytokine staining, 1x 10
6
splenocytes were plated into
a 96-well plate. Cells were suspended and incubated in RPMI 1640 culture medium and stimu-
lated for four hours utilizing phorbol 12-myristate 13-acetate (30ng/mL) and ionomycin
(400ng/mL) with 10μg/mL of Brefeldin A at 37°C. After stimulation, the cells were stained with
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 4/20
anti-CD4 PacBlue (BD Bioscience), anti-CD8 PacOrange (Life Technologies Carlsbad, CA),
anti-IL-2 FITC (BD Bioscience), anti-IL-4 PE (eBioscience), anti-CD44 PerCP (BioLegend),
anti-CCR4 PeCy7 (BioLegend), anti-CXCR3 APC (eBioscience), and anti-IFNy Alexa700 (BD
Bioscience). An LSR II flow cytometer (BD Biosciences) was used to run all samples and FlowJo
10.0.8r1 software (Tree Star, San Carlos, CA) was used to analyze all data.
Intestinal permeability
At 19 hours following CLP, mice were gavaged with 0.5 ml of fluorescein isothiocyanate-dex-
tran (FD4) at a concentration of 22mg/ml in PBS (Sigma-Aldrich)[21,22]. Plasma collected at
time of sacrifice 5 hour later was then diluted with an equal volume of PBS, and FD4 concen-
tration was determined by flourospectrometry with excitation/emission wavelengths of 485/
520 nm with a standard curve of serial dilutions (BioTex Synergy HT, Winooski, VT).
Intestinal proliferation
Proliferating intestinal epithelial cells were stained with 5-Bromo-2deoxyuridine (BrdU). At
90 minutes prior to sacrifice, mice received intraperitoneal injections of BrdU (5mg/ml in 0.9%
saline, Sigma-Aldrich) to label S-phase cells [23,24]. Jejunal tissue was then fixed in 10% forma-
lin for 24 hours before being embedded in paraffin and slide-mounted in 5μm sections. Slides
were then deparaffinized, rehydrated, and incubated in 1% hydrogen peroxide for 15 minutes
before being heated in a pressure cooker in antigen decloaker (Biocare Medical, Concord, CA)
for 45 minutes. Protein block (Dako, Carpinteria, CA) was performed for 30 minutes at room
temperature and slides were incubated overnight at 4°C with rat monoclonal Anti-BrdU
(1:500; Accurate Chemical and Scientific, Westbury, NJ). Samples were then incubated with
goat anti-rat antibody (1:500; Accurate Chemical & Scientific) and streptavidin horseradish
peroxidase (1:500; Dako), each for an hour at room temperature. Diaminobenzidine (DAB)
was used to develop slides for 23 minutes, and counterstaining was performed with hematox-
ylin. BrdU-positive cells were quantified in 100 contiguous, well-oriented intestinal crypts.
Intestinal apoptosis
Apoptosis of intestinal epithelial cells was quantified using two complementary techniques:
active caspase-3 staining and morphologic analysis of hematoxylin-eosin stained sections
[25,26]. Sections were treated as above with antigen decloaker and were then blocked with 20%
normal goat serum (Vector Laboratories, Burlingame, CA). Slides were incubated overnight at
4°C with rabbit anti-caspase-3 (1:100; Cell Signaling, Beverly, MA), and then with goat anti-
rabbit biotinylated antibody (1:500; Vector Laboratories) and streptavidin horseradish peroxi-
dase (1:500; Dako) for one hour each at room temperature. Slides were developed with DAB
and then counterstained. Caspase-3 positive cells were counted in 100 contiguous intestinal
crypts.
Apoptotic cells were identified on hematoxylin-eosin-stained sections by identifying charac-
teristic morphological changes including cell shrinkage with condensed and fragmented nuclei
and quantifying them in 100 contiguous crypts.
Villus length
Villus length was measured as the distance in μm from the crypt neck to the villus tip in 12 con-
secutive well-oriented jejunal villi using Nikon Elements imaging software- EIS-Elements BR
3.10 (Nikon Instruments, Melville, NY).
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 5/20
Bacterial cultures
Quantitative cultures of whole blood and peritoneal fluid were prepared from serial 10-fold
dilutions of samples in sterile 0.9% saline. A 100 μl aliquot of undiluted sample and each dilu-
tion from 10
1
to 10
3
was plated on blood agar plates (Remel, Lenexa, KS) and incubated at
35°C in a 5% CO
2
atmosphere for 24 hours. Colony counts were obtained from plates contain-
ing fewer than 300 colonies. The number of colony-forming units (CFUs) per ml of original
sample was determined by multiplying the number of colonies by the reciprocal of the dilution
counted and adjusted for the volume of sample plated.
Cytokines
Whole blood, peritoneal fluid and bronchoalveolar lavage (BAL) fluid were collected and then
centrifuged at 10,000 RPM for 10 minutes. The supernatant from each was then collected and
analyzed for cytokine concentrations using a 6-plex cytokine bead array according to manufac-
turer instructions (Bio-Rad Laboratories, Hercules, CA).
Renal function
Whole blood was centrifuged at 10,000 RPM for 10 minutes. Serum creatinine was then mea-
sured using a creatinine microplate assay (Oxford Biomedical Research, Rochester Hills, MI)
while blood urea nitrogen (BUN) was determined using a urea nitrogen colorimetric detection
kit (Arbor Assays, Ann Arbor, MI). Whole kidneys were removed and fixed in 10% formalin
for 24 hours before paraffin fixation and sectioning. After hematoxylin-eosin staining, sections
were assessed for injury by a pathologist blinded to sample identity (ABF). Animal weights
were measured at time of CLP and immediately prior to sacrifice to assess potential weight loss
due to dehydration.
Liver injury
Liver injury was evaluated by both serum liver enzymes and histology. Alanine aminotransfer-
ase (ALT) was measured on a Beckman AU480 chemistry auto-analyzer (Beckman Diagnos-
tics, LaBrea, CA) following manufacturer instructions. In addition, portions of whole liver
were removed and fixed in 10% formalin for 24 hours prior to paraffin embedding. Sections
were then slide-mounted and stained with hematoxylin-eosin for analysis by a pathologist
(ABF) blinded to sample identity.
Lung injury
For histologic analysis, lungs of animals were flushed with 1ml of 10% formalin, and sections
were then removed and fixed in 10% formalin for 24 hours prior to being embedded in paraf-
fin. Lung sections were then stained with hematoxylin-eosin and examined by a pathologist
(ABF) blinded to sample identity.
Myeloperoxidase (MPO) activity was also assessed in BAL fluid. The trachea was irrigated
with 1 ml of PBS, and fluid was then withdrawn and centrifuged at 10,000 RPM for 10 minutes.
Substrate buffer containing 0.0005% hydrogen peroxide and O-dianisidine was added to the
supernatant, and MPO activity was assayed over 6 minutes at wavelength 460 (BioTek Synergy
HT, Winooski, VT). MPO activity was calculated as optical density/minute per μl of BAL fluid.
Lung fluid protein concentration was measured by analyzing samples treated with protein
assay reagent (Thermo Scientific, Rockford, IL) at 660 nm in conjunction with a standard
curve of bovine serum albumin.
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 6/20
Complete blood counts
Whole blood collected at time of sacrifice was collected in anti-coagulant-lined blood tubes and
was analyzed for hemoglobin concentration, leukocyte count, and platelet count on a Heska
HemaTrue
1
veterinary hematology analyzer per manufacturer guidelines (Heska, Loveland, CO).
Statistics
All data were analyzed using the statistical software program Prism 6.0 (GraphPad, San Diego,
CA) and are presented as mean ± SEM. Data were tested for Gaussian distribution using the
D'Agostino-Pearson omnibus normality test. Two -way comparisons on data with a Gaussian
distribution were performed using the Students t-test. Two-way comparisons on data that did
not have a Gaussian distribution were performed using the Mann-Whitney test. Multi-group
comparisons were analyzed via one-way ANOVA, followed by the Tukey post-test. Survival
was analyzed using the Log-Rank test. A p value of <0.05 was considered to be statistically sig-
nificant throughout.
Results
Mice that received subcutaneous injections of murine lung cancer cells developed well-circum-
scribed solitary tumors at the site of injection three weeks later (average size 1.5 cm in diame-
ter). Post-mortem review of lung histology demonstrated microscopic metastatic disease in
some animals, although no gross tumor spread was seen in any animals at time of sacrifice.
The presence of pre-existing lung cancer worsens mortality following
sepsis
Seven-day mortality was 18% in previously healthy septic mice. In contrast, seven-day mortal-
ity was 60% in cancer septic mice (Fig 1).
The presence of pre-existing lung cancer decreases CD4+ lymphocytes
but not CD8+ lymphocytes following sepsis
Cancer septic mice had a decrease in both the frequency and absolute numbers of splenic
CD4+ lymphocytes compared to previously healthy septic mice (Fig 2A and 2B). In addition,
Fig 1. Effect of cancer on survival from sepsis. Previously healthy mice and those given an injection of
LLC1 cells three weeks earlier (n = 15-17/group) were subjected to CLP. Cancer septic mice had significantly
higher mortality than previously healthy septic mice (p = 0.003).
doi:10.1371/journal.pone.0149069.g001
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 7/20
cancer septic mice had a higher frequency of annexin-positive staining CD4+ lymphocytes
compared to previously healthy septic mice, suggesting increased apoptosis was responsible for
the loss of CD4+ cells (Fig 2C and 2D). In contrast, while the frequency of splenic CD8+ lym-
phocytes was higher in cancer septic mice (Fig 3A), this was likely due to the decrease in CD4
+ cells since no difference was seen in CD8+ cell numbers (Fig 3B) or annexin staining (Fig 3C
and 3D) between cancer septic mice and previously healthy septic mice. CD4+ cell expression
of the Th1 marker CXCR3 was increased in cancer septic mice (Fig 4A and 4C), while expres-
sion of the Th2 marker CCR4 was not different between previously healthy septic mice and
cancer septic mice (Fig 4B and 4C). Stimulated production of Th1 effector cytokine IFN-γby
CD4+ T cells was not impacted by cancer (Fig 4D, 4F and 4G) while production of the Th2
effector IL-4 was significantly decreased in cancer septic mice (Fig 4E, 4F and 4G).
The presence of pre-existing lung cancer decreases ability to clear local
infection
Peritoneal bacterial burden was higher in cancer septic mice than previously healthy septic mice
(Fig 5A). This was not associated with changes in Interleukin (IL)-1β, IL-6, IL-10, IL-13, MCP-1,
Fig 2. Effect of cancer on splenic CD4+ T cells following sepsis. Cancer septic mice had a significantly lower frequency of CD4+ T cells as a percentage
of total CD3+ T cells (A, p = 0.02, n= 79) as well as a decrease in the total number ofCD4+ T cells (B, p = 0.03, n = 810) compared to previously healthy
septic mice. This was associated withan increase in annexin-positive CD4+ T cells in cancer septicmice (C, p = 0.04, n = 78). A representative flow cytometry
histogram demonstrates increased annexin staining in cancer septic (blue) CD4+ T cells compared to previously healthy septic (red) CD4+ T cells (D).
doi:10.1371/journal.pone.0149069.g002
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 8/20
TNF-α,orIFN-γin the peritoneal fluid (Fig 5B5H). Peritoneal fluid also contained similar per-
centages of neutrophils (Fig 6A) and dendritic cells (Fig 6B) between previously healthy septic
mice and cancer septic mice. Dendritic cell activation as determined by MHC II expression on
cells from peritoneal fluid was also similar between the groups (Fig 6C). Splenic dendritic cell fre-
quency and activation were also unaffected by the presence of cancer (Fig 6D and 6E). There
were no differences in absolute numbers of neutrophils or dendritic cells (data not shown).
Serum cytokines were similar between cancer septic mice and previously healthy septic
mice except for an increase in MCP-1 in cancer septic mice (Fig 7). No difference in bacterial
burden was identified in quantitative blood cultures between previously healthy septic mice
and cancer septic mice (data not shown).
The presence of pre-existing lung cancer decreases crypt proliferation
but does not alter other components of intestinal integrity following sepsis
Cancer in isolation decreases crypt proliferation compared to unmanipulated mice. Sepsis in
isolation also decreases crypt proliferation compared to unmanipulated mice. The combination
Fig 3. Effect of cancer on splenic CD8+ T cells following sepsis. Cancer septic mice had a significantly increased frequency of CD8+ T cells as a
percentage of total CD3+ T cells (A, p = 0.007, n = 810). However, there was no statistically significant change in total number of CD8+ cells (B, p = 0.10,
n=810) or annexin-positive CD8+ T cells (C, p = 0.31, n = 710) suggesting the change in percentage of CD8+ cells was related to the decrease in CD4
+ cells. A representative flow cytometry histogram demonstrates no difference in annexin staining in cancer septic (blue) and previously healthy septic (red)
CD8+ T cells (D).
doi:10.1371/journal.pone.0149069.g003
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 9/20
of cancer and sepsis causes a further disproportionate decrease in crypt proliferation compared
to sepsis alone as cancer septic mice have lower proliferation than previously healthy septic
mice (Fig 8A8C).
In contrast, villus length is not affected by cancer in isolation, is decreased as has been previ-
ously shown by sepsis (26), but is not different between previously healthy septic mice and can-
cer septic mice (Fig 8D). Cancer and sepsis also did not impact intestinal permeability or crypt
apoptosis compared to sepsis alone (data not shown).
The presence of pre-existing lung cancer worsens biochemical kidney
function without causing liver injury following sepsis
Neither cancer nor sepsis in isolation affected renal function as BUN and Cr levels were similar
in unmanipulated, cancer and previously healthy septic mice. In contrast, both BUN and Cr
were higher in cancer septic mice (Fig 9A and 9B); however kidneys appeared grossly normally
Fig 4. Effect of cancer on Th1 and Th2 markers and cytokine production following sepsis. Cancer septic mice had a modest increase in CD4+ T cell
expression of the Th1 marker CXCR3 (A, p = 0.01, n = 910) but did not have a statistically significant change in expression of the Th2 marker CCR4 (B,
p = 0.21, n = 910). A representative flow cytometry plot for both markers is included (C). Stimulated production of IFN-γwas not statistically different in CD4
+ T cells from cancer septic mice (D, p = 0.17, n = 910); however, stimulated production of IL-4 was lower in CD4+ T cells from cancer septic mice (E,
p = 0.006, n = 910). Representative flow cytometry plots for both unstimulated and stimulated IFN-γand IL-4 are shown for previously healthy septic mice
(F) and cancer septic mice (G).
doi:10.1371/journal.pone.0149069.g004
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 10 / 20
histologically (data not shown). No differences in body weight were identified between previ-
ously healthy septic mice and cancer septic mice (Fig 9C).
Basal levels of ALT were similar in unmanipulated and cancer mice. While sepsis increased
serum ALT levels, this was not affected by the presence of cancer since ALT was similar
between cancer septic and previously healthy septic mice (Fig 9D). Further, there were no dif-
ferences in liver histology between any of the groups (data not shown).
The presence of pre-existing lung cancer decreases BAL MPO and
increases BAL protein following sepsis
MPO activity was lower in BAL fluid in cancer septic mice compared to previously healthy sep-
tic mice (Fig 10A). In contrast, BAL protein was higher in cancer septic mice compared to
previously healthy septic mice (Fig 10B). BAL cytokines were generally independent of the
presence of cancer although BAL IL-10 was lower in cancer septic mice (Fig 10C10G). Despite
differences noted in BAL fluid, no differences were identified in inflammation score or percent-
age of inflammatory cells between cancer septic and previously healthy septic mice on histo-
logic examination (data not shown).
Fig 5. Effect of cancer on peritoneal bacteria and local inflammatory response following sepsis. Cancer septic mice had higher levels of bacteria in
their peritoneal cavities than previously healthy septic mice (A, p = 0.005, n = 89). This change was not associated with differences in concentration of
peritoneal IL-1β(p = 0.32), IL-6 (p = 0.18), IL-10 (p = 0.52), IL-13 (p = 0.97), MCP-1 (p = 0.67), TNF-α(p = 0.40), or IFN-γ(p = 0.27) (B-H, n = 8 for all groups).
doi:10.1371/journal.pone.0149069.g005
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 11 / 20
Fig 6. Effect of cancer on local neutrophil and dendritic cell responses following sepsis. Cancer septic mice and previously healthy septic mice had
similar frequencies of neutrophils (A, p = 0.52, n = 7/group) and dendritic cells (B, p = 0.52, n = 7/group)in their peritoneal fluid, and there was no difference in
the frequency of dendritic cell MHC II expression between the two groups (C, p = 0.52, n = 7/group). Cancer septic mice and previously healthy septic mice
also had similar frequencies of dendritic cells (D, p = 0.73, n = 910) and activated dendritic cells (E, p = 0.83, n = 910) in splenocytes.
doi:10.1371/journal.pone.0149069.g006
Fig 7. Effect of cancer on systemic cytokines following sepsis. No significant differences were noted in IL-1β(p = 0.10), IL-6 (p = 0.63), IL-10 (p = 0.24),
IL-13 (p = 0.07), or TNF-α(p = 0.99) (B-F, n = 7-8/group). In contrast, increased MCP-1 was detected in the serum of cancer septic mice compared to
previously healthy septic mice (E, p = 0.04, n = 78).
doi:10.1371/journal.pone.0149069.g007
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 12 / 20
The presence of pre-existing lung cancer does not alter complete blood
counts following sepsis
Cancer, in isolation, causes anemia as hemoglobin levels were significantly lower in cancer
mice than unmanipulated mice (Fig 11A). However, sepsis, in isolation did not alter hemoglo-
bin levels and there was no statistically significant difference in hemoglobin levels between pre-
viously healthy septic mice and cancer septic mice. Cancer, in isolation, did not change total
leukocyte count although sepsis, in isolation, decreased total leukocyte count (Fig 11B). The
combination of sepsis and cancer did not alter total leukocyte count further since it was similar
Fig 8. Effect of cancer on intestinal proliferation following sepsis. Previously healthy septic mice (A) had qualitatively higher levels of proliferation than
cancer septic mice (B, BrdU-positive crypt cells stain brown). Quantitatively, both cancer in isolation and sepsis in isolation decrease crypt proliferation (C,
p = 0.02 and 0.008 respectively compared to unmanipulated mice). The combination of cancer and sepsis further decreased intestinal proliferation, moreso
than was seen with either variable in isolation (p<0.001 previously healthy septic vs. cancer septic, n = 810 for all groups in panel C). In contrast, while villus
length was decreased by sepsis (but not cancer) in isolation (p = 0.008), there was no difference in villus length between previously healthy septic and cancer
septic mice (D, p>0.99, n = 89 for all groups).
doi:10.1371/journal.pone.0149069.g008
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 13 / 20
between previously healthy septic mice and cancer septic mice. Neither cancer nor sepsis in iso-
lation led to a statistically significant change in platelet count compared to unmanipulated ani-
mals, and similarly, there were no statistically significant differences in platelet count between
previously healthy septic mice and cancer septic mice (Fig 11C).
Fig 9. Effect of cancer on renal and liver function following sepsis. Neither cancer nor sepsis in isolation impacted BUN or Cr levels (A, B). However, the
combination of both insults worsens renal function as both BUN and Cr were higher in cancer septic mice than previously healthy septic mice (p = 0.004 and
p<0.0001 respectively, n = 8 for all groups). Body weights were similar in all groups, regardlessof the presence of cancer or a septic insult 24 hours earlier
(C). While cancer had no impact on liver function as assayed by ALT, sepsis, in isolation, increased ALT compared to unmanipulated mice (D, p<0.001).
However, this was not worsened by malignancy as cancer septic mice and previously healthy septic mice had similar ALT levels (p>0.99, n = 810 for all
groups).
doi:10.1371/journal.pone.0149069.g009
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 14 / 20
Discussion
Similar to the human condition and our previous mouse model of cancer followed by sepsis,
this study demonstrated that the presence of a malignancy significantly worsens survival from
sepsis. However, parameters associated with increased mortality varied greatly between mice
injected with lung cancer cells followed by CLP in this study and our prior findings of mice
injected with pancreatic cancer cells followed by Pseudomonas aeruginosa pneumonia. In this
study, we noted increased apoptosis in splenic CD4+ T lymphocytes, decreased crypt prolifera-
tion, decreased local infection clearance, increased systemic MCP-1, worse renal function,
lower BAL MPO activity, higher BAL protein and lower BAL IL-10 in cancer septic mice com-
pared to previously healthy septic mice. In contrast, in mice with pancreatic cancer cells fol-
lowed by Pseudomonas aeruginosa pneumonia, we found decreased apoptosis in both T and B
lymphocytes, increased gut epithelial apoptosis, increased bacteremia without alterations in
local infection, and higher BAL IL-6 and IL-10 compared to previously healthy septic mice.
Remarkably, except for elevated mortality, there was essentially no overlap in the associated
abnormalities between lung cancer/CLP and pancreatic cancer/pneumonia mice.
Fig 10. Effect of cancer on lung inflammation following sepsis. MPO activity in BAL fluid was decreased in cancer septic mice compared to previously
healthy septic mice (A, p<0.0001, n = 8). In contrast, protein concentration in BAL fluid was higher in cancer septic mice (B, p = 0.04, n = 68). BAL cytokines
were similar between mice for IL-1β(p = 0.47), IL-6 (p = 0.59), IFN-γ(p = 0.99) and TNF-α(p = 0.11) but IL-10 levels were lower in cancer septic mice
(p = 0.0005, n = 910 for all groups).
doi:10.1371/journal.pone.0149069.g010
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 15 / 20
Whether a common host response exists in sepsis is controversial [2729]. Our results can
be interpreted that the mechanisms responsible for mortality vary significantly, at least in part,
with tumor type and sepsis model. Alternatively, since our results are associative, it is possible
that none of the abnormalities detected in either model of cancer and sepsis is actually respon-
sible for the increased mortality. While we cannot rule out that possibility, it is reasonable to
attempt to put the findings described herein into context of existing literature.
The concept that T lymphocyte derangements play a crucial role in mediating mortality
from sepsis is now over a decade old. Multiple studies demonstrate that preventing T cell apo-
ptosis improves survival following CLP [3032]. The findings that a) CD4+ lymphocytes are
decreased and b) Annexin V staining is increased are consistent with a more profound immu-
nosuppressive state in mice with lung cancer followed by CLP compared to previously healthy
mice subjected to the same insult. While this would typically be thought of as maladaptive, it
should be noted that prevention of lymphocyte apoptosis worsened survival in mice overex-
pressing Bcl-2 in lymphocytes as well as in BIM knockouts following pneumonia in mice with
pancreatic cancer [15], so the functional significance of increased CD4+ lymphocyte apoptosis
in cancer septic mice needs to be examined further in future experiments.
Altered Th1 and Th2 responses have also been described in sepsis [17,33], and it seemed
plausible that cancer-mediated increases in T cell exhaustion could potentially induce a shift
towards a Th2 response during sepsis [12]. However, our results do not neatly fit this hypothe-
sis. Although we demonstrated a modest increase in expression of CXCR3 on CD4+ T cells in
cancer septic mice (which may suggest a skewing towards a Th1 response), the biological sig-
nificance of this is unclear since we found no associated differences in production of the Th1
effector IFN-γby CD4+ T cells. In addition, we did not detect a difference in expression of the
Th2 marker CCR4 but did observe decreased production of the Th2 effector IL-4 from CD4
+ cells taken from cancer septic mice. These somewhat conflicting data suggest that while there
may be imbalances in the Th1/Th2 response following sepsis in animals with cancer, the
changes are likely not a simple shift directly toward one phenotype or another, and the degree
to which those changes impact mortality is yet to be determined.
Fig 11. Effect of cancer on hemoglobin, leukocyte count and platelet count following sepsis. Cancer, in isolation, decreased serum hemoglobin levels
(A, p<0.0001); however, this was not impacted by sepsis and there was no statistically significant difference between cancer septic and previously healthy
septic mice (p = 0.14, n = 5-9/group). In contrast, while cancer did not impact leukocyte count (B), sepsis, in isolation caused a decrease in in leukocyte count
(p = 0.02). No statistically significant difference in leukocyte count was seen between cancer septic mice and previously healthy mice (p = 0.99, n = 5-9/
group). Platelet count was similar between unmanipulated mice and those with cancer or sepsis in isolation (C). No statistically significant difference in
platelet count was seen between cancer septic mice and previously healthy mice (p = 0.78, n = 5-9/group).
doi:10.1371/journal.pone.0149069.g011
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 16 / 20
There were significantly higher levels of bacteria present in the peritoneal cavity 24 hours
after CLP in cancer septic mice. Even though local cytokine, neutrophil, and dendritic cell pro-
files were similar between cancer septic and previously healthy septic mice, it is plausible that
the basal immunosuppressive state caused by sepsis predisposes animals to increased local
infection. How this suppression is functionally enacted requires further investigation in future
studies. Notably, polymicrobial sepsis, as would typically be seen following fecal peritonitis, is
associated with higher mortality in patients with cancer and sepsis[9].
Gut integrity is altered by sepsis, with increased apoptosis and permeability as well as
decreased proliferation and villus length. These and other alterations in gut integrity can result
in distant organ injury and propagation of systemic inflammation, resulting in the hypothesis
that the gut is the "motor" driving critical illness [16,34,35]. Although multiple parameters of
gut integrity were altered by sepsis in isolation in this study, the majority of these were similar
between cancer septic mice and previously healthy septic mice. One exception was gut prolifer-
ation, which was decreased by both cancer and sepsis in isolation but disproportionately
decreased by the combination of both insults. In light of the complex function and architecture
of the gut, it is possible that the marked decrease in gut proliferation seen in cancer septic mice
resulted in downstream functional changes within the intestine, resulting in either local or dis-
tant injury. Since the gut is a continuously renewing organ that replaces itself on average every
35 days[35], future experiments are required to determine how long the decreased prolifera-
tion induced by the combination of sepsis and cancer persists.
Both BUN and Cr (markers of renal dysfunction) are similar in unmanipulated mice, cancer
mice and previously healthy septic mice, suggesting neither cancer nor sepsis impacts renal
function. However, BUN and Cr are statistically higher in cancer septic mice compared to previ-
ously healthy septic mice. We do not believe that the modest degree of biochemical acute kidney
injury seen at 24 hours is solely responsible for mortality seen in cancer septic mice, and it is
questionable if the small absolute difference in serum BUN/creatinine values is biologically
meaningful, especially given the absence of histologic abnormalities in the kidneys in all groups.
It is possible that renal function worsens throughout the course of sepsis in this model and
therefore contributes to mortality more significantly at later time points. Of note, the etiology of
renal dysfunction that is seen exclusively in cancer septic mice remains to be determined.
Finally, MPO activity, protein and IL-10 were all altered in BAL fluid of cancer septic mice
although this was not accompanied by differences in histologic lung inflammation or other
BAL cytokines between previously healthy septic mice and cancer septic mice. While lowered
MPO activity and decreased levels IL-10 may suggest a dampened pulmonary inflammatory
response, more detailed assays would be needed to determine if this is actually the case. In
addition, since studies have shown that pulmonary disease does not represent a significant
cause of death in mice subjected to CLP[36], the functional significance of these findings is
unclear.
This study has a number of limitations. While a number of abnormalities were identified
that are associated with increased mortality, we cannot conclude that any of them are causative
without performing additional mechanistic studies. Next, all non-survival studies were per-
formed at a single timepoint (24 hours), so it is likely that our experimental design missed tem-
poral trends that would be important towards understanding the relationship between cancer
and sepsis, especially since there appear to be different inflammatory states depending on how
far out a host is from their septic insult [37]. Next, although we have attempted to compare our
results to our previous study on pancreatic cancer/pneumonia, there are three variables that
are different in this manuscripttype of cancer, sepsis model and the fact that micro-meta-
static disease was noted in this study with LLC-1 cells whereas Pan02 pancreatic cancer cells
are not associated with metastatic disease. To determine which of these are most responsible
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 17 / 20
for differences, changing only a single variable at a time would be required. Next, tumor cells
were injected into the thigh of murine recipients and thus do not replicate the development of
an in-situ lung cancer. Mice with naturally occurring tumors could potentially be exposed to
local tumor-related factors that cause a different inflammatory milieu than is seen in our
model. Further a more gradual tumor onset growth could potentially alter dynamics of tumor-
immune cell interaction, leading to chronic inflammatory changes not seen after only three
weeks of tumor growth. Finally, tumors tend to develop in aged patients, whereas this study
examined mice that were 912 weeks old prior to the onset of sepsis. It is unclear how well
young mice function as surrogates for aged patients, and our experimental design precluded us
from assessing the impact of age on the pathophysiology of cancer and sepsis.
Despite these limitations, our data yield new insights into a clinically relevant model of both
a common cancer and a common cause of sepsis, and the interplay between the two insults.
Further research is required to determine if the multiple pathophysiologic abnormalities identi-
fied herein are important in mediating the increased mortality seen when sepsis occurs in the
setting of cancer, and the role of specific tumors or types of sepsis in mediating this complex
interaction.
Author Contributions
Conceived and designed the experiments: JDL MLF CMC. Performed the experiments: JDL
RM ZL LMM EMB ABF KF CWC. Analyzed the data: JDL RM ZL LMM EMB ABF MLF CMC
KF CWC. Contributed reagents/materials/analysis tools: EMB ABF MLF CMC. Wrote the
paper: JDL CMC. Revised the manuscript: RM ZL LMM EMB ABF MLF KF CWC.
References
1. Gaieski DF, Edwards JM, Kallan MJ, Carr BG. Benchmarking the incidence and mortality of severe
sepsis in the United States. Crit Care Med 2013 May; 41(5):116774. doi: 10.1097/CCM.
0b013e31827c09f8 PMID: 23442987
2. Danai PA, Moss M, Mannino DM, Martin GS. The epidemiology of sepsis in patients with malignancy.
Chest 2006 June; 129(6):143240. PMID: 16778259
3. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe
sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care
Med 2001 July; 29(7):130310. PMID: 11445675
4. Melamed A, Sorvillo FJ. The burden of sepsis-associated mortality in the United States from 1999to
2005: an analysis of multiple-cause-of-death data. Crit Care 2009; 13(1):R28. doi: 10.1186/cc7733
PMID: 19250547
5. Williams MD, Braun LA, Cooper LM, Johnston J, Weiss RV, Qualy RL, et al. Hospitalized cancer
patients with severe sepsis: analysis of incidence, mortality, and associated costs of care. Crit Care
2004 October; 8(5):R291R298. PMID: 15469571
6. Soares M, Caruso P, Silva E, Teles JM, Lobo SM, Friedman G, et al. Characteristics and outcomes of
patients with cancer requiring admission to intensive care units: a prospective multicenter study. Crit
Care Med 2010 January; 38(1):915. doi: 10.1097/CCM.0b013e3181c0349e PMID: 19829101
7. Rosolem MM, Rabello LS, Lisboa T, Caruso P, Costa RT, Leal JV, et al. Critically ill patients with cancer
and sepsis: clinical course and prognostic factors. J Crit Care 2012 June; 27(3):3017. doi: 10.1016/j.
jcrc.2011.06.014 PMID: 21855281
8. de ME, Tandjaoui-Lambiotte Y, Legrand M, Lambert J, Mokart D, Kouatchet A, et al. Outcomes in criti-
cally ill cancer patients with septic shock of pulmonary origin. Shock 2013 March; 39(3):2504. doi: 10.
1097/SHK.0b013e3182866d32 PMID: 23364436
9. Torres VB, Azevedo LC, Silva UV, Caruso P, Torelly AP, Silva E, et al. Sepsis-Associated Outcomes in
Critically Ill Patients with Malignancies. Ann Am Thorac Soc 2015 Aug; 12(8):118592. doi: 10.1513/
AnnalsATS.201501-046OC PMID: 26086679
10. Safdar A, Armstrong D. Infectious morbidity in critically ill patients with cancer. Crit Care Clin 2001 July;
17(3):531viii. PMID: 11525048
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 18 / 20
11. Mittal R, Chen CW, Lyons JD, Margoles LM, Liang Z, Coopersmith CM, et al. Murine lung cancer
induces generalized T-cell exhaustion. J Surg Res 2015 May 15; 195(2):5419. doi: 10.1016/j.jss.
2015.02.004 PMID: 25748104
12. Mittal R, Wagener M, Breed ER, Liang Z, Yoseph BP, Burd EM, et al. Phenotypic T cell exhaustion in a
murine model of bacterial infection in the setting of pre-existing malignancy. PLoS ONE 2014; 9(5):
e93523. doi: 10.1371/journal.pone.0093523 PMID: 24796533
13. Hotchkiss RS, Moldawer LL. Parallels between cancer and infectious disease. N Engl J Med 2014 July
24; 371(4):3803. doi: 10.1056/NEJMcibr1404664 PMID: 25054723
14. Fox AC, Robertson CM, Belt B, Clark AT, Chang KC, Leathersich AM, et al. Cancer causes increased
mortality and is associated with altered apoptosis in murine sepsis. Crit Care Med 2010 March; 38
(3):88693. doi: 10.1097/CCM.0b013e3181c8fdb1 PMID: 20009755
15. Fox AC, Breed ER, Liang Z, Clark AT, Zee-Cheng BR, Chang KC, et al. Prevention of Lymphocyte Apo-
ptosis in Septic Mice with Cancer Increases Mortality. J Immunol 2011 August 15; 187(4):19506. doi:
10.4049/jimmunol.1003391 PMID: 21734077
16. Mittal R, Coopersmith CM. Redefining the gut as the motor of critical illness. Trends Mol Med 2014
April; 20(4):21423. doi: 10.1016/j.molmed.2013.08.004 PMID: 24055446
17. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions
to immunotherapy. Nat Rev Immunol 2013 December; 13(12):86274. doi: 10.1038/nri3552 PMID:
24232462
18. Wiersinga WJ, Leopold SJ, Cranendonk DR, van der Poll T. Host innate immune responses to sepsis.
Virulence 2014 January 1; 5(1):3644. doi: 10.4161/viru.25436 PMID: 23774844
19. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, et al. Surviving Sepsis Campaign:
International Guidelines for Management of Severe Sepsis and Septic Shock: 2012. Crit Care Med
2013 February; 41(2):580637. doi: 10.1097/CCM.0b013e31827e83af PMID: 23353941
20. Baker CC, Chaudry IH, Gaines HO, Baue AE. Evaluation of factors affecting mortality rate after sepsis
in a murine cecal ligation and puncture model. Surgery 1983 August; 94(2):3315. PMID: 6879447
21. Yoseph BP, Breed E, Overgaard CE, Ward CJ, Liang Z, Wagener ME, et al. Chronic alcohol ingestion
increases mortality and organ injury in a murine model of septic peritonitis. PLoSONE 2013; 8(5):
e62792. doi: 10.1371/journal.pone.0062792 PMID: 23717394
22. Clark JA, Gan H, Samocha AJ, Fox AC, Buchman TG, Coopersmith CM. Enterocyte-specific epidermal
growth factor prevents barrier dysfunction and improves mortality in murine peritonitis. Am J Physiol
Gastrointest Liver Physiol 2009 September; 297(3):G471G479. doi: 10.1152/ajpgi.00012.2009 PMID:
19571236
23. Liang Z, Xie Y, Dominguez JA, Breed ER, Yoseph BP, Burd EM, et al. Intestine-specific deletion of
microsomal triglyceride transfer protein increases mortality in aged mice. PLoS ONE 2014; 9(7):
e101828. doi: 10.1371/journal.pone.0101828 PMID: 25010671
24. Dominguez JA, Xie Y, Dunne WM, Yoseph BP, Burd EM, Coopersmith CM, et al. Intestine-specific
Mttp deletion decreases mortality and prevents sepsis-induced intestinal injury in a murine model of
Pseudomonas aeruginosa pneumonia. PLoS ONE 2012; 7(11):e49159. doi: 10.1371/journal.pone.
0049159 PMID: 23145105
25. Vyas D, Robertson CM, Stromberg PE, Martin JR, Dunne WM, Houchen CW, et al. Epithelial apoptosis
in mechanistically distinct methods of injury in the murine small intestine. Histol Histopathol 2007 June;
22(6):62330. PMID: 17357092
26. Dominguez JA, Samocha AJ, Liang Z, Burd EM, Farris AB, Coopersmith CM. Inhibition of IKKbeta in
Enterocytes Exacerbates Sepsis-Induced Intestinal Injury and Worsens Mortality. Crit Care Med 2013
October; 41(10):e275e285. doi: 10.1097/CCM.0b013e31828a44ed PMID: 23939348
27. McConnell KW, McDunn JE, Clark AT, Dunne WM, Dixon DJ, Turnbull IR, et al. Streptococcus pneu-
moniae and Pseudomonas aeruginosa pneumonia induce distinct host responses. Crit Care Med 2010
January; 38(1):22341. doi: 10.1097/CCM.0b013e3181b4a76b PMID: 19770740
28. Fry DE. The generic septic response. Crit Care Med 2008 April; 36(4):136970. doi: 10.1097/CCM.
0b013e31816a11e9 PMID: 18379273
29. Yu SL, Chen HW, Yang PC, Peck K, Tsai MH, Chen JJ, et al. Differential gene expression in gram-neg-
ative and gram-positive sepsis. Am J Respir Crit Care Med 2004 May 15; 169(10):113543. PMID:
15001460
30. Hotchkiss RS, Tinsley KW, Swanson PE, Chang KC, Cobb JP, Buchman TG, et al. Prevention of lym-
phocyte cell death in sepsis improves survival in mice. Proc Natl Acad Sci U S A 1999 December 7; 96
(25):145416. PMID: 10588741
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 19 / 20
31. Hotchkiss RS, Swanson PE, Knudson CM, Chang KC, Cobb JP, Osborne DF, et al. Overexpression of
Bcl-2 in transgenic mice decreases apoptosis and improves survival in sepsis.J Immunol 1999 April 1;
162(7):414856. PMID: 10201940
32. Hotchkiss RS, Coopersmith CM, McDunn JE, Ferguson TA. The sepsis seesaw: tilting toward immuno-
suppression. Nat Med 2009 May; 15(5):4967. doi: 10.1038/nm0509-496 PMID: 19424209
33. Delano MJ, Scumpia PO, Weinstein JS, Coco D, Nagaraj S, Kelly-Scumpia KM, et al. MyD88-depen-
dent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polar-
ization in sepsis. J Exp Med 2007 June 11; 204(6):146374. PMID: 17548519
34. Sertaridou E, Papaioannou V, Kolios G, Pneumatikos I. Gut failure in critical care: old school versus
new school. Ann Gastroenterol 2015 July; 28(3):30922. PMID: 26130136
35. Clark JA, Coopersmith CM. Intestinal crosstalk: a new paradigm for understanding the gut as the
"motor" of critical illness. Shock 2007 October; 28(4):38493. PMID: 17577136
36. Iskander KN, Craciun FL, Stepien DM, Duffy ER, Kim J, Moitra R,et al. Cecal ligation and puncture-
induced murine sepsis does not cause lung injury. Crit Care Med 2013 January; 41(1):15465.
37. Hotchkiss RS, Sherwood ER. Immunology. Getting sepsis therapy right. Science 2015 March 13; 347
(6227):12012. doi: 10.1126/science.aaa8334 PMID: 25766219
Murine Lung Cancer and Sepsis
PLOS ONE | DOI:10.1371/journal.pone.0149069 March 28, 2016 20 / 20
... The majority of cancer patients have active/progressing tumors at the time of sepsis and experience greater sepsis-associated mortality than patients without cancer (2,4). These clinical outcomes have been reverse-translated in murine models using multiple sepsis models (e.g., cecal ligation and puncture [CLP]) and malignant cell types (e.g., pancreatic adenocarcinoma) (5)(6)(7). Improved treatment has increased survival of the entire septic population and has greatly benefitted cancer patients, resulting in a 76% survival rate after sepsis (2). Thus, cancer patients have high incidence of sepsis, and it is important to understand how sepsis impacts cancer prognosis and longterm outcomes of an increasing number of sepsis survivors. ...
... Experimental data in mice using various sepsis and/or tumor models have recapitulated the increased rate of sepsis-induced mortality in cancer patients (5)(6)(7). To independently confirm and further develop this concept, B6 mice were injected with B16 cells s.c. ...
Article
Full-text available
Malignancy increases sepsis incidence 10-fold and elevates sepsis-associated mortality. Advances in treatment have improved survival of cancer patients shortly after sepsis, but there is a paucity of information on how sepsis impacts cancer growth, development, and prognosis. To test this, cecal ligation and puncture surgery was performed on B16 melanoma-bearing mice to show that sepsis has detrimental effects in hosts with advanced tumors, leading to increased mortality. Surprisingly, mice experiencing cecal ligation and puncture-induced sepsis earlier during tumor development exhibited CD8 T cell-dependent attenuation of tumor growth. Sepsis-resistant CD8 tumor-infiltrating T cells showed increased in vivo activation, effector IFN-γ cytokine production, proliferation, and expression of activation/inhibitory PD-1/LAG-3 receptors because of a sepsis-induced liberation of tumor Ags. Sepsis-reinvigorated CD8 tumor-infiltrating T cells were also amenable to (anti-PD-L1/LAG-3) checkpoint blockade therapy, further prolonging cancer-associated survival in sepsis survivors. Thus, sepsis has the capacity to improve tumor-specific CD8 T cell responses, leading to better cancer prognosis and increased survival.
... Because cancer-septic mice have a higher mortality compared with septic mice without cancer (21,22), we next assessed whether tumor-specific T cells contributed to this increase in sepsis mortality. To do this, anti-Thy1.1 Ab (or isotype control) was administered to deplete OT-I cells 3 wk following adoptive transfer of Thy1.1 1 OT-I and LLC-OVA tumor induction. ...
Article
Full-text available
Sepsis induces significant immune dysregulation characterized by lymphocyte apoptosis and alterations in the cytokine milieu. Because cancer patients exhibit a 10-fold greater risk of developing sepsis compared with the general population, we aimed to understand how pre-existing malignancy alters sepsis-induced immune dysregulation. To address this question, we assessed the impact of tumor-specific CD8+ T cells on the immune response in a mouse model of cecal ligation and puncture (CLP)-induced sepsis. Tumor-bearing animals containing Thy1.1+ tumor-specific CD8+ T cells were subjected to CLP, and groups of animals received anti-Thy1.1 mAb to deplete tumor-specific CD8+ T cells or isotype control. Results indicated that depleting tumor-specific T cells significantly improved mortality from sepsis. The presence of tumor-specific CD8+ T cells resulted in increased expression of the 2B4 coinhibitory receptor and increased apoptosis of endogenous CD8+ T cells. Moreover, tumor-specific T cells were not reduced in number in the tumors during sepsis but did exhibit impaired IFN-γ production in the tumor, tumor draining lymph node, and spleen 24 h after CLP. Our research provides novel insight into the mechanisms by which pre-existing malignancy contributes to increased mortality during sepsis.
... Tumor-induced expansion of MDSCs has been identified as a factor of susceptibility to both endotoxinic shock and polymicrobial peritonitis in relation with cachexia, lung and liver infiltration by myeloid cells, and increased VȮ 2 (55). Mice subjected to subcutaneous tumor graft of pancreatic adenocarcinoma displayed defective bacterial clearance and hypersusceptibility to Pseudomonas aeruginosa pneumonia and polymicrobial peritonitis (56,57). Lymphocyte apoptosis, an immune hallmark of sepsis, has been consistently associated with mortality and is liable to antiapoptotic strategies to improve survival in experimental models. ...
Article
Sepsis and cancer share a number of pathophysiological features and both result from the inability of the host's immune system to cope with the initial insult, respectively tissue invasion by pathogens or malignant cell transformation. The common coexistence of both disorders and the profound related alterations in immune homeostasis raise the question of their mutual impact on each other's course. This translational review aims at discussing the interactions between cancer and sepsis supported by clinical data and the translation to experimental models. The dramatic improvement in cancer has come at a cost of increased risks of life-threatening infectious complications. Investigating the long-term outcome of sepsis survivors has revealed an unexpected susceptibility to cancer long after discharge from intensive care unit. Nonetheless it is noteworthy that an acute septic episode may harbor anti-tumoral properties under particular circumstances. Relevant double-hit animal models have provided clues to whether and how a bacterial sepsis may impact on malignant tumor growth. In sequential sepsis-then-cancer models, post-septic mice exhibited accelerated tumor growth. When using reverse cancer-then-sepsis models, bacterial sepsis applied to mice with cancer conversely resulted in inhibition or even regression of tumor growth. Experimental models thus highlight dual effects of sepsis on tumor growth mostly depending on the sequence of insults, and allow deciphering the immune mechanisms and their relation with microorganisms.
... MLCK activation is commonly found with bacterial infection [35,36], and inhibition of MLCK improves survival in a mouse model of sepsis [37] as well as improving barrier function and tight junction rearrangement in a murine model of burn injury [38]. Of note, changes to the gut epithelium and barrier function are exacerbated in the presence of chronic co-morbidities such as cancer [39,40] or chronic alcohol use [41][42][43]. ...
Article
Full-text available
Background The gut is hypothesized to be the “motor” of critical illness. Under basal conditions, the gut plays a crucial role in the maintenance of health. However, in critical illness, all elements of the gut are injured, potentially worsening multiple organ dysfunction syndrome. Main body Under basal conditions, the intestinal epithelium absorbs nutrients and plays a critical role as the first-line protection against pathogenic microbes and as the central coordinator of mucosal immunity. In contrast, each element of the gut is impacted in critical illness. In the epithelium, apoptosis increases, proliferation decreases, and migration slows. In addition, gut barrier function is worsened via alterations to the tight junction, resulting in intestinal hyperpermeability. This is associated with damage to the mucus that separates the contents of the intestinal lumen from the epithelium. Finally, the microbiome of the intestine is converted into a pathobiome, with an increase in disease-promoting bacteria and induction of virulence factors in commensal bacteria. Toxic factors can then leave the intestine via both portal blood flow and mesenteric lymph to cause distant organ damage. Conclusion The gut plays a complex role in both health and critical illness. Here, we review gut integrity in both health and illness and highlight potential strategies for targeting the intestine for therapeutic gain in the intensive care unit.
... An extensive literature exists on the importance of apoptosis-mostly occurring in rapidly dividing cells such as lymphocytes and the gut epithelium-in sepsis. 5,6 However, much less is known about the role of other forms of cell death. Pyroptosis is a lytic form of programmed cell death mediated by inflammatory caspases and characterized by the formation of large pores on the plasma membrane. ...
Article
Sepsis is one of the leading causes of death worldwide. While mortality is high regardless of inciting infection or comorbidities, mortality in patients with cancer and sepsis is significantly higher than mortality in patients with sepsis without cancer. Cancer patients are also significantly more likely to develop sepsis than the general population. The mechanisms underlying increased mortality in cancer and sepsis patients are multifactorial. Cancer treatment alters the host immune response and can increase susceptibility to infection. Preclinical data also suggests that cancer, in and of itself, increases mortality from sepsis with dysregulation of the adaptive immune system playing a key role. Further, preclinical data demonstrate that sepsis can alter subsequent tumor growth while tumoral immunity impacts survival from sepsis. Checkpoint inhibition is a well-accepted treatment for many types of cancer, and there is increasing evidence suggesting this may be a useful strategy in sepsis as well. However, preclinical studies of checkpoint inhibition in cancer and sepsis demonstrate results that could not have been predicted by examining either variable in isolation. As sepsis management transitions from a 'one size fits all' model to a more individualized approach, understanding the mechanistic impact of cancer on outcomes from sepsis represents an important strategy towards delivering on the promise of precision medicine in the intensive care unit.
Article
Full-text available
Sepsis is a leading cause of morbidity and mortality associated with significant impairment in memory T cells. These changes include the upregulation of co-inhibitory markers, a decrease in functionality, and an increase in apoptosis. Due to recent studies describing IL-27 regulation of TIGIT and PD-1, we assessed whether IL-27 impacts these co-inhibitory molecules in sepsis. Based on these data, we hypothesized that IL-27 was responsible for T cell dysfunction during sepsis. Using the cecal ligation and puncture (CLP) sepsis model, we found that IL-27Rα was associated with the upregulation of TIGIT on memory CD4⁺ T cells following CLP. However, IL-27 was not associated with sepsis mortality.
Article
Full-text available
Numerous models are available for the preclinical study of sepsis, and they fall into one of three general categories: (1) administration of exogenous toxins (e.g., lipopolysaccharide, zymosan), (2) virulent bacterial or viral challenge, and (3) host barrier disruption, e.g., cecal ligation and puncture (CLP) or colon ascendens stent peritonitis (CASP). Of the murine models used to study the pathophysiology of sepsis, CLP combines tissue necrosis and polymicrobial sepsis secondary to autologous fecal leakage, as well as hemodynamic and biochemical responses similar to those seen in septic humans. Further, a transient numerical reduction of multiple immune cell types, followed by development of prolonged immunoparalysis, occurs in CLP‐induced sepsis just as in humans. Use of the CLP model has led to a vast expansion in knowledge regarding the intricate physiological and cellular changes that occur during and after a septic event. This updated article details the steps necessary to perform this survival surgical technique, as well as some of the obstacles that may arise when evaluating the sepsis‐induced changes within the immune system. It also provides representative monoclonal antibody (mAb) panels for multiparameter flow cytometric analysis of the murine immune system in the septic host. © 2020 Wiley Periodicals LLC. Basic Protocol : Cecal ligation and puncture in the mouse
Article
Objectives: Our goal was to "reverse translate" the human response to surgical sepsis into the mouse by modifying a widely adopted murine intra-abdominal sepsis model to engender a phenotype that conforms to current sepsis definitions and follows the most recent expert recommendations for animal preclinical sepsis research. Furthermore, we aimed to create a model that allows the study of aging on the long-term host response to sepsis. Design: Experimental study. Setting: Research laboratory. Subjects: Young (3-5 mo) and old (18-22 mo) C57BL/6j mice. Interventions: Mice received no intervention or were subjected to polymicrobial sepsis with cecal ligation and puncture followed by fluid resuscitation, analgesia, and antibiotics. Subsets of mice received daily chronic stress after cecal ligation and puncture for 14 days. Additionally, modifications were made to ensure that "Minimum Quality Threshold in Pre-Clinical Sepsis Studies" recommendations were followed. Measurements and main results: Old mice exhibited increased mortality following both cecal ligation and puncture and cecal ligation and puncture + daily chronic stress when compared with young mice. Old mice developed marked hepatic and/or renal dysfunction, supported by elevations in plasma aspartate aminotransferase, blood urea nitrogen, and creatinine, 8 and 24 hours following cecal ligation and puncture. Similar to human sepsis, old mice demonstrated low-grade systemic inflammation 14 days after cecal ligation and puncture + daily chronic stress and evidence of immunosuppression, as determined by increased serum concentrations of multiple pro- and anti-inflammatory cytokines and chemokines when compared with young septic mice. In addition, old mice demonstrated expansion of myeloid-derived suppressor cell populations and sustained weight loss following cecal ligation and puncture + daily chronic stress, again similar to the human condition. Conclusions: The results indicate that this murine cecal ligation and puncture + daily chronic stress model of surgical sepsis in old mice adhered to current Minimum Quality Threshold in Pre-Clinical Sepsis Studies guidelines and met Sepsis-3 criteria. In addition, it effectively created a state of persistent inflammation, immunosuppression, and weight loss, thought to be a key aspect of chronic sepsis pathobiology and increasingly more prevalent after human sepsis.
Article
Patients with cancer who develop sepsis have a markedly higher mortality than patients who were healthy prior to the onset of sepsis. Potential mechanisms underlying this difference have previously been examined in two preclinical models of cancer followed by sepsis. Both pancreatic cancer/pneumonia and lung cancer/cecal ligation and puncture (CLP) increase murine mortality, associated with alterations in lymphocyte apoptosis and intestinal integrity. However, pancreatic cancer/pneumonia decreases lymphocyte apoptosis and increases gut apoptosis while lung cancer/CLP increases lymphocyte apoptosis and decreases intestinal proliferation. These results cannot distinguish the individual roles of cancer versus sepsis since different models of each were used. We therefore created a new cancer/sepsis model to standardize each variable. Mice were injected with a pancreatic cancer cell line and three weeks later cancer mice and healthy mice were subjected to CLP. Cancer septic mice had a significantly higher 10-day mortality than previously healthy septic mice. Cancer septic mice had increased CD4 T cells and CD8 T cells, associated with decreased CD4 T cell apoptosis 24 hours after CLP. Further, splenic CD8+ T cell activation was decreased in cancer septic mice. In contrast, no differences were noted in intestinal apoptosis, proliferation or permeability, nor were changes noted in local bacterial burden, renal, liver or pulmonary injury. Cancer septic mice thus have consistently reduced survival compared to previously healthy septic mice, independent of the cancer or sepsis model utilized. Changes in lymphocyte apoptosis are common to cancer model and independent of sepsis model whereas gut apoptosis is common to sepsis model and independent of cancer model. The host response to the combination of cancer and sepsis is dependent, at least in part, on both chronic co-morbidity and acute illness.
Article
Full-text available
Sepsis is a major cause of mortality for critically ill patients with cancer. Information about clinical outcomes and factors associated with increased risk of death in these patients are necessary to help physicians recognize those patients who are most likely to benefit from ICU therapy and identify possible targets for intervention. In this study, we evaluated cancer patients with sepsis from a multicenter prospective study in order to characterize their clinical characteristics and to identify independent risk factors associated with hospital mortality. Subgroup analysis of a multicenter prospective cohort study conducted in 28 Brazilian intensive care units (ICUs) evaluating adult cancer patients with severe sepsis and septic shock. We used logistic regression to identify variables associated with hospital mortality. Out of the 717 patients admitted to the participating ICUs, 268 (37%) had severe sepsis (n=142, 53%) or septic shock (n=126, 47%) and comprised the population of this study. Mean SAPS 3 score was 62.9±17.7 points and median SOFA score was 9 (7-12) points, respectively. The most frequent sites of infection were the lungs (48%), intra-abdominal (25%), primary bloodstream infections (19%) and urinary tract (17%). Half of patients had microbiologically proven infections and Gram-negative bacteria were the most common pathogens causing sepsis (31%). ICU and hospital mortality rates were 42% and 56%, respectively. In multivariable analysis, the number of acute organ dysfunctions [odds-ratio (OR)=1.48(95% CI, 1.16-1.87)], hematological malignancy [OR=2.57 (1.05-6.27)], performance status 2-4 [OR=2.53(1.44-4.43)] and polymicrobial infections [OR=3.74 (1.52-9.21)] were associated with hospital mortality. Sepsis is a common cause of critical illness in cancer patients and remains associated with high mortality. Variables related to underlying malignancy, sepsis severity and characteristics of infection are associated with grim prognosis.
Article
Full-text available
The concept of bacterial translocation and gut-origin sepsis as causes of systemic infectious complications and multiple organ deficiency syndrome in surgical and critically ill patients has been a recurring issue over the last decades attracting the scientific interest. Although gastrointestinal dysfunction seemingly arises frequently in intensive care unit patients, it is usually underdiagnosed or underestimated, because the pathophysiology involved is incompletely understood and its exact clinical relevance still remains controversial with an unknown yet probably adverse impact on the patients' outcome. The purpose of this review is to define gut-origin sepsis and related terms, to describe the mechanisms leading to gut-derived complications, and to illustrate the therapeutic options to prevent or limit these untoward processes.
Article
Full-text available
Background Mice with conditional, intestine-specific deletion of microsomal triglyceride transfer protein (Mttp-IKO) exhibit a complete block in chylomicron assembly together with lipid malabsorption. Young (8–10 week) Mttp-IKO mice have improved survival when subjected to a murine model of Pseudomonas aeruginosa-induced sepsis. However, 80% of deaths in sepsis occur in patients over age 65. The purpose of this study was to determine whether age impacts outcome in Mttp-IKO mice subjected to sepsis. Methods Aged (20–24 months) Mttp-IKO mice and WT mice underwent intratracheal injection with P. aeruginosa. Mice were either sacrificed 24 hours post-operatively for mechanistic studies or followed seven days for survival. Results In contrast to young septic Mttp-IKO mice, aged septic Mttp-IKO mice had a significantly higher mortality than aged septic WT mice (80% vs. 39%, p = 0.005). Aged septic Mttp-IKO mice exhibited increased gut epithelial apoptosis, increased jejunal Bax/Bcl-2 and Bax/Bcl-XL ratios yet simultaneously demonstrated increased crypt proliferation and villus length. Aged septic Mttp-IKO mice also manifested increased pulmonary myeloperoxidase levels, suggesting increased neutrophil infiltration, as well as decreased systemic TNFα compared to aged septic WT mice. Conclusions Blocking intestinal chylomicron secretion alters mortality following sepsis in an age-dependent manner. Increases in gut apoptosis and pulmonary neutrophil infiltration, and decreased systemic TNFα represent potential mechanisms for why intestine-specific Mttp deletion is beneficial in young septic mice but harmful in aged mice as each of these parameters are altered differently in young and aged septic WT and Mttp-IKO mice.
Article
Full-text available
While much of cancer immunology research has focused on anti-tumor immunity both systemically and within the tumor microenvironment, little is known about the impact of pre-existing malignancy on pathogen-specific immune responses. Here, we sought to characterize the antigen-specific CD8+ T cell response following a bacterial infection in the setting of pre-existing pancreatic adenocarcinoma. Mice with established subcutaneous pancreatic adenocarcinomas were infected with Listeria monocytogenes, and antigen-specific CD8+ T cell responses were compared to those in control mice without cancer. While the kinetics and magnitude of antigen-specific CD8+ T cell expansion and accumulation was comparable between the cancer and non-cancer groups, bacterial antigen-specific CD8+ T cells and total CD4+ and CD8+ T cells in cancer mice exhibited increased expression of the coinhibitory receptors BTLA, PD-1, and 2B4. Furthermore, increased inhibitory receptor expression was associated with reduced IFN-γ and increased IL-2 production by bacterial antigen-specific CD8+ T cells in the cancer group. Taken together, these data suggest that cancer's immune suppressive effects are not limited to the tumor microenvironment, but that pre-existing malignancy induces phenotypic exhaustion in T cells by increasing expression of coinhibitory receptors and may impair pathogen-specific CD8+ T cell functionality and differentiation.
Article
Full-text available
Sepsis - which is a severe life-threatening infection with organ dysfunction - initiates a complex interplay of host pro-inflammatory and anti-inflammatory processes. Sepsis can be considered a race to the death between the pathogens and the host immune system, and it is the proper balance between the often competing pro- and anti-inflammatory pathways that determines the fate of the individual. Although the field of sepsis research has witnessed the failure of many highly touted clinical trials, a better understanding of the pathophysiological basis of the disorder and the mechanisms responsible for the associated pro- and anti-inflammatory responses provides a novel approach for treating this highly lethal condition. Biomarker-guided immunotherapy that is administered to patients at the proper immune phase of sepsis is potentially a major advance in the treatment of sepsis and in the field of infectious disease.
Article
Objective To provide an update to the original Surviving Sepsis Campaign clinical management guidelines, “Surviving Sepsis Campaign guidelines for management of severe sepsis and septic shock,” published in 2004.
Article
Objective: To provide an update to the "Surviving Sepsis Campaign Guidelines for Management of Severe Sepsis and Septic Shock," last published in 2008. Design: A consensus committee of 68 international experts representing 30 international organizations was convened. Nominal groups were assembled at key international meetings (for those committee members attending the conference). A formal conflict of interest policy was developed at the onset of the process and enforced throughout. The entire guidelines process was conducted independent of any industry funding. A stand-alone meeting was held for all subgroup heads, co- and vice-chairs, and selected individuals. Teleconferences and electronic-based discussion among subgroups and among the entire committee served as an integral part of the development. Methods: The authors were advised to follow the principles of the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system to guide assessment of quality of evidence from high (A) to very low (D) and to determine the strength of recommendations as strong (1) or weak (2). The potential drawbacks of making strong recommendations in the presence of low-quality evidence were emphasized. Some recommendations were ungraded (UG). Recommendations were classified into three groups: 1) those directly targeting severe sepsis; 2) those targeting general care of the critically ill patient and considered high priority in severe sepsis; and 3) pediatric considerations. Results: Key recommendations and suggestions, listed by category, include: early quantitative resuscitation of the septic patient during the first 6 hrs after recognition (1C); blood cultures before antibiotic therapy (1C); imaging studies performed promptly to confirm a potential source of infection (UG); administration of broad-spectrum antimicrobials therapy within 1 hr of recognition of septic shock (1B) and severe sepsis without septic shock (1C) as the goal of therapy; reassessment of antimicrobial therapy daily for de-escalation, when appropriate (1B); infection source control with attention to the balance of risks and benefits of the chosen method within 12 hrs of diagnosis (1C); initial fluid resuscitation with crystalloid (1B) and consideration of the addition of albumin in patients who continue to require substantial amounts of crystalloid to maintain adequate mean arterial pressure (2C) and the avoidance of hetastarch formulations (1C); initial fluid challenge in patients with sepsis-induced tissue hypoperfusion and suspicion of hypovolemia to achieve a minimum of 30 mL/kg of crystalloids (more rapid administration and greater amounts of fluid may be needed in some patients) (1C); fluid challenge technique continued as long as hemodynamic improvement, as based on either dynamic or static variables (UG); norepinephrine as the first-choice vasopressor to maintain mean arterial pressure ≥ 65 mm Hg (1B); epinephrine when an additional agent is needed to maintain adequate blood pressure (2B); vasopressin (0.03 U/min) can be added to norepinephrine to either raise mean arterial pressure to target or to decrease norepinephrine dose but should not be used as the initial vasopressor (UG); dopamine is not recommended except in highly selected circumstances (2C); dobutamine infusion administered or added to vasopressor in the presence of a) myocardial dysfunction as suggested by elevated cardiac filling pressures and low cardiac output, or b) ongoing signs of hypoperfusion despite achieving adequate intravascular volume and adequate mean arterial pressure (1C); avoiding use of intravenous hydrocortisone in adult septic shock patients if adequate fluid resuscitation and vasopressor therapy are able to restore hemodynamic stability (2C); hemoglobin target of 7-9 g/dL in the absence of tissue hypoperfusion, ischemic coronary artery disease, or acute hemorrhage (1B); low tidal volume (1A) and limitation of inspiratory plateau pressure (1B) for acute respiratory distress syndrome (ARDS); application of at least a minimal amount of positive end-expiratory pressure (PEEP) in ARDS (1B); higher rather than lower level of PEEP for patients with sepsis-induced moderate or severe ARDS (2C); recruitment maneuvers in sepsis patients with severe refractory hypoxemia due to ARDS (2C); prone positioning in sepsis-induced ARDS patients with a PaO2/FIO2 ratio of ≤ 100 mm Hg in facilities that have experience with such practices (2C); head-of-bed elevation in mechanically ventilated patients unless contraindicated (1B); a conservative fluid strategy for patients with established ARDS who do not have evidence of tissue hypoperfusion (1C); protocols for weaning and sedation (1A); minimizing use of either intermittent bolus sedation or continuous infusion sedation targeting specific titration endpoints (1B); avoidance of neuromuscular blockers if possible in the septic patient without ARDS (1C); a short course of neuromuscular blocker (no longer than 48 hrs) for patients with early ARDS and a Pao2/Fio2 < 150 mm Hg (2C); a protocolized approach to blood glucose management commencing insulin dosing when two consecutive blood glucose levels are > 180 mg/dL, targeting an upper blood glucose ≤ 180 mg/dL (1A); equivalency of continuous veno-venous hemofiltration or intermittent hemodialysis (2B); prophylaxis for deep vein thrombosis (1B); use of stress ulcer prophylaxis to prevent upper gastrointestinal bleeding in patients with bleeding risk factors (1B); oral or enteral (if necessary) feedings, as tolerated, rather than either complete fasting or provision of only intravenous glucose within the first 48 hrs after a diagnosis of severe sepsis/septic shock (2C); and addressing goals of care, including treatment plans and end-of-life planning (as appropriate) (1B), as early as feasible, but within 72 hrs of intensive care unit admission (2C). Recommendations specific to pediatric severe sepsis include: therapy with face mask oxygen, high flow nasal cannula oxygen, or nasopharyngeal continuous PEEP in the presence of respiratory distress and hypoxemia (2C), use of physical examination therapeutic endpoints such as capillary refill (2C); for septic shock associated with hypovolemia, the use of crystalloids or albumin to deliver a bolus of 20 mL/kg of crystalloids (or albumin equivalent) over 5 to 10 mins (2C); more common use of inotropes and vasodilators for low cardiac output septic shock associated with elevated systemic vascular resistance (2C); and use of hydrocortisone only in children with suspected or proven "absolute"' adrenal insufficiency (2C). Conclusions: Strong agreement existed among a large cohort of international experts regarding many level 1 recommendations for the best care of patients with severe sepsis. Although a significant number of aspects of care have relatively weak support, evidence-based recommendations regarding the acute management of sepsis and septic shock are the foundation of improved outcomes for this important group of critically ill patients.
Article
Sepsis—a complication of infection—is a factor in at least a third of all hospital deaths—a sobering statistic ( 1 ). Patients with sepsis frequently present with fever, shock, and multiorgan failure. Because of this dramatic clinical scenario, investigators have generally assumed that sepsis mortality is due to unbridled inflammation ( 2 ). Research in animal models, in which administration of the cytokines tumor necrosis factor–α (TNF-α) and interleukin-1 (IL-1) reproduced many features of sepsis, supported that assertion. Yet, over 40 clinical trials of agents that block cytokines, pathogen recognition, or inflammation-signaling pathways have universally failed ( 3 , 4 ). However, on page 1260 of this issue, Weber et al. ( 5 ) show that blocking a cytokine—specifically, IL-3—can indeed be protective against sepsis.
Article
Cancer is known to modulate tumor-specific immune responses by establishing a microenvironment that leads to the upregulation of T-cell inhibitory receptors, resulting in the progressive loss of function and eventual death of tumor-specific T-cells. However, the ability of cancer to impact the functionality of the immune system on a systemic level is much less well characterized. Because cancer is known to predispose patients to infectious complications including sepsis, we hypothesized that the presence of cancer alters pathogen-directed immune responses on a systemic level. We assessed systemic T-cell coinhibitory receptor expression, cytokine production, and apoptosis in mice with established subcutaneous lung cancer tumors and in unmanipulated mice without cancer. Results indicated that the frequencies of programmed death-1-positive, B and T lymphocyte attenuator-positive, and 2B4(+) cells in both the CD4(+) and CD8(+) T-cell compartments were increased in mice with localized cancer relative to non-cancer controls, and the frequencies of both CD4(+) and CD8(+) T-cells expressing multiple different inhibitory receptors were increased in cancer animals relative to non-cancer controls. Additionally, 2B4(+)CD8(+) T-cells in cancer mice exhibited reduced interleukin-2 and interferon-γ, whereas B and T lymphocyte attenuator-positive CD8(+) T-cells in cancer mice exhibited reduced interleukin-2 and tumor necrosis factor. Conversely, CD4(+) T-cells in cancer animals demonstrated an increase in the frequency of annexin V(+) apoptotic cells. Taken together, these data suggest that the presence of cancer induces systemic T-cell exhaustion and generalized immune suppression. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
The status of the immune system affects and is affected by both cancer and chronic infection. Some molecular mechanisms of immunity are relevant to both disease states.