Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Intestine-Specific Deletion of Microsomal Triglyceride Transfer Protein Increases Mortality in Aged Mice

  • Zhe Liang,

    Affiliation Emory Center for Critical Care and Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, United States of America

  • Yan Xie,

    Affiliation Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Jessica A. Dominguez,

    Affiliation Department of Basic Sciences, Bastyr University California, San Diego, California, United States of America

  • Elise R. Breed,

    Affiliation Emory Center for Critical Care and Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, United States of America

  • Benyam P. Yoseph,

    Affiliation Emory Center for Critical Care and Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, United States of America

  • Eileen M. Burd,

    Affiliation Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America

  • Alton B. Farris,

    Affiliation Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America

  • Nicholas O. Davidson ,

    Contributed equally to this work with: Nicholas O. Davidson, Craig M. Coopersmith

    Affiliation Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Craig M. Coopersmith

    Contributed equally to this work with: Nicholas O. Davidson, Craig M. Coopersmith

    cmcoop3@emory.edu

    Affiliation Emory Center for Critical Care and Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, United States of America

Abstract

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.

Introduction

Sepsis affects over 800,000 patients annually in the United States, and between 230,000 and 370,000 patients die of the disease annually [1]. Sepsis is a disease of the aged. Even though elderly patients account for slightly greater than 10% of the population in the United States, more than 60% of cases of sepsis and approximately 80% of deaths occur in patients over 65 years of age [2], [3]. Both animal and human studies demonstrate that age greatly impacts the host response following sepsis and not only is mortality higher with aging, but also multiple therapies that are effective in young mice are ineffective in aged mice [4][12]. Despite this, the vast majority of rodent models of sepsis use 6–16 week old animals, which corresponds to a 10–17 year old human [13]. It has been suggested that the use of young animals to model a disease of the aged – which have altered inflammation, coagulation, apoptosis, and proteomics – is one of many reasons why preclinical models of sepsis have failed to translate into positive clinical trials [14], [15].

The gut has long been hypothesized to be “the motor” of the systemic inflammatory response syndrome [16][19]. Critical illness worsens gut integrity in a number of ways, including increasing intestinal epithelial apoptosis and permeability while simultaneously decreasing both proliferation and mucous production [20][44]. The microbiome of the gut – which ordinarily exists in a symbiotic relationship with the host – can also respond to environmental cues in sepsis to induce virulence factors, which can, in turn, worsen critical illness [45][48]. Additionally, toxic gut-derived lymph induces distant organ injury in critical illness as numerous lines of evidence support the “gut lymph” hypothesis in which the intestine releases toxic mediators that are transported through the mesenteric lymph nodes and cause damage remote from the intestine [49][57].

The role of intestinal chylomicron assembly and secretion in transporting toxic gut-derived lipid factors is poorly understood. Microsomal triglyceride transfer protein (Mttp) resides within the lumen of the endoplasmic reticulum and plays a requisite role in the assembly of triglyceride-rich lipoprotein particles, both within enterocytes of the small intestine and also in hepatocytes of the liver [58]. We previously studied the impact of sepsis in young (8–10 week old) mice with defective chylomicron assembly induced by conditional intestine-specific deletion of Mttp (Mttp-IKO mice) [59]. Compared to young septic wild type (WT) mice, young septic Mttp-IKO mice have increased survival. This is associated with improvements in gut integrity, manifested by decreased gut apoptosis, increased crypt proliferation and longer villus length. Young septic Mttp-IKO mice also have marked decreases in systemic IL-6 and G-CSF. Despite pneumonia being initiated in the lungs, no significant pulmonary differences exist between young septic Mttp-IKO and WT mice, with similar levels of MPO activity, bacterial clearance and broncoalveolar lavage (BAL) cytokines. The aim of this study was to determine if aging alters the beneficial impact seen with intestine-specific Mttp deletion in young septic mice.

Materials and Methods

Animals

Aged (defined as 20–24 months old) Mttpflox/flox villin-Cre-ERT2 (Mttp-IKO) mice on a background of ∼75% C57BL/6 and ∼25% 129/SvJ were utilized for all experiments. Conditional, intestine-specific deletion of Mttp was induced via Cre recombinase expression in villus epithelial cells in which animals received five daily injections of tamoxifen given intraperitoneally (1 mg, Sigma, St. Louis, MO) [59], [60]. Experiments were performed three weeks after tamoxifen injections. Age matched littermate animals that did not receive tamoxifen (i.e. functionally WT) were used as controls. All 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-2000539-121812). All surgery (described below) was performed under isoflurane anesthesia and all efforts were made to minimize animal suffering by treating all animals with buprenex post-operatively. For non-survival studies, animals were sacrificed 24 hours following intratracheal injection of P. aeruginosa (detailed below) using asphyxiation by CO2 or via exsanguination under deep isoflurane anesthesia. A different subset of animals was followed for 7 days after intratracheal injection of P. aeruginosa for survival studies. Animals were checked twice daily. Moribund animals were sacrificed using humane endpoints. The following criteria were used to identify moribund animals: a) major organ failure or medical conditions unresponsive to treatment such as severe respiratory distress, icterus, uremia, intractable diarrhea, or self-mutilation, b) surgical complications unresponsive to immediate intervention (bleeding, infection, wound dehiscence) or c) clinical or behavioral signs unresponsive to appropriate intervention persisting for 24 hours including significant inactivity, labored breathing, sunken eyes, hunched posture, piloerection/matted fur, one or more unresolving skin ulcers, and abnormal vocalization when handled. Note that it is important that many classical humane endpoints for animal studies are not sufficient for studies in sepsis, as many animals meeting general guidelines criteria for euthanasia survive despite sepsis. Specific guidelines for humane endpoints in sepsis research [61], [62] were followed. Non-moribund animals that died during the course of the survival curve died as a direct result of their septic insult. Animals that survived 7 days after intratracheal injection of P. aeruginosa were sacrificed at the conclusion of this experiment using asphyxiation by CO2.

Sepsis model

Under isoflurane anesthesia, all animals were subjected to intratracheal injections of P. aeruginosa (ATCC 27853) via midline cervical incision [63], [64]. A total of 40 µl of bacteria diluted in normal saline (2–4×107 CFU) was injected via a 29 gauge syringe. To enhance bacterial delivery, mice were held vertically for 10 seconds. All mice received a subcutaneous injection of 1 ml saline post-operatively to compensate for insensible fluid losses. Animals were either euthanized 24 hours post-operatively or followed 7 days for survival.

Intestinal morphology

Villus length was measured as the distance in µm from the crypt neck to the villus tip. Crypt depth was measured as the distance in µm from the crypt base to crypt neck. Each was measured in 12–16 well-oriented jejunal villi and crypts using Nikon Elements imaging software- EIS-Elements BR 3.10 (Nikon Instruments, Melville, NY).

Intracellular lipid droplets were evaluated using osmium tetoxide staining in intestinal tissue fixed in 10% neutral buffered formalin. Sections were then transferred into 1% osmium tetroxide, rinsed in distilled water, incubated in 0.5% periodic acid, washed and then processed for paraffin embedding, followed by counterstaining with hematoxylin and eosin (H&E).

Intestinal apoptosis

Apoptotic cells were quantified in the jejunum using two independent but complementary techniques: active caspase-3 staining and morphologic analysis of H&E-stained sections [27]. For caspase-3 staining, sections were deparaffinized, rehydrated, and incubated in 3% hydrogen peroxide for 10 minutes. Sections were then placed in Antigen Decloaker (Biocare Medical, Concord, CA) and heated in a pressure cooker for 45 minutes. After sections were blocked with 20% normal goat serum (Vector Laboratories, Burlingame, CA), they were incubated overnight with rabbit polyclonal anti-active caspase-3 (1∶100; Cell Signaling, Beverly, MA) at 4°C. The following day, sections were incubated with goat anti-rabbit biotinylated secondary antibody (1∶200; Accurate Chemical and Scientific, Westbury, NJ) for 1 hour followed by horseradish peroxidase (HRP)- labeled streptavidin (Accurate Chemical and Scientific) for 1 hour. Sections were then developed with diaminobenzidine and counterstained with hematoxylin.

Apoptotic cells were identified on H&E-stained sections via characteristic morphological changes including cell shrinkage with condensed and fragmented nuclei and then quantified in 100 well-oriented contiguous crypt-villus units.

Intestinal proliferation

Mice were intraperitoneally injected with 5-Bromo-2′deoxyuridine (BrdU, 5 mg/ml diluted in normal saline; Sigma) 90 minutes prior to sacrifice to label cells in S-phase. Sections were deparaffinized, rehydrated, incubated in 1% hydrogen peroxide for 15 minutes, immersed in Antigen Decloaker (Biocare Medical) and heated in a pressure cooker for 45 minutes. Sections were then blocked with Protein Block (Dako, Carpinteria, CA) for 10 minutes, and incubated with rat monoclonal anti-BrdU overnight at 4°C (1∶500; Accurate Chemical & Scientific). The following day, sections were incubated with goat anti-rat biotinylated secondary antibody (1∶500; Accurate Chemical and Scientific) for 1 hour, followed by HRP labeled Streptavidin (Accurate Chemical and Scientific) for 1 hour and then developed with diaminobenzidine and counterstained with hematoxylin. BrdU-stained cells were quantified in 100 well-oriented contiguous crypts.

Gene expression of apoptosis mediators

Total RNA was isolated from frozen jejunal tissue using the RNeasy Mini Kit (QIAGEN, Santa Clarita, CA) according to manufacturer protocol [59]. RNA integrity was verified by electrophoresis and cDNA was synthesized from 0.5 µg of total RNA. Bax, Bcl-2, and Bcl-xL mRNA levels were detected using pre-developed TaqMan primers and probes (Applied Biosystems, Foster City, CA) and run on the ABI StepOnePlus Real-Time PCR system (Applied Biosystems). Samples were run in duplicate and normalized to expression of the endogenous control, glyceraldehyde-3-phosphate. Relative quantification of PCR products were based upon value differences between the target gene and glyceraldehyde-3-phosphate using the comparative CT method.

Bacterial cultures

Bronchoalveolar lavage (BAL) fluid was obtained by cannulating the trachea and lavaging the lungs with 1 ml of sterile saline. BAL samples were serially diluted in sterile saline and plated on sheep’s blood agar plates. Plates were incubated overnight at 37°C in 5% CO2 and colony counts were determined 24 hours later. From plates containing fewer than 300 colonies.

Myeloperoxidase (MPO) activity

BAL fluid was centrifuged at 5,000 rpm for 5 minutes. Following addition of substrate buffer containing O-dianisidine and 0.0005% hydrogen peroxide, MPO activity was measured at 460 nm wavelengths over 6 minutes (Bio-Tek Instruments-Synergy HT, Winooski, VT). MPO activity was calculated as optical density/minute (U) per µl of BAL fluid.

Systemic and BAL cytokines

Blood was collected at time of sacrifice, centrifuged at 5,000 rpm for 5 minutes in serum separator tubes and stored at −80°C until use. Both serum and BAL levels of IL-1β, IL-6, IL-10, IL-13, MCP-1, and TNFα were measured by using a multiplex cytokine assay (Bio-Rad) according to manufacturer’s instructions [29]. All samples were run in duplicate.

Serum and tissue lipids

Blood was collected on the same animals immediately prior to induction of pneumonia and 24 hours later. Serum trigylcerides (TG) and cholesterol levels were determined using commercially available kits (Wako Chemicals, Richmond, VA). Lipid quantitation in the proximal jejunum was performed as previously described [59]. Cholesterol and triglyceride distribution within high density lipoproteins (HDL) and low density lipoproteins (LDL) were quantified following fractionation using fast protein liquid chromatography using tandem Superose 6 columns [65]. Serum bile acid levels were determined using an enzymatic assay kit (Cat No. BQ-092A-EALD, BioQuatnt, San Diego, CA)according to manufacturer’s instructions.

Liver and splenic pathology

H&E-stained section of liver and spleen were evaluated by a pathologist blinded to sample identity (ABF).

Statistical analysis

Continuous data sets were tested for Gaussian distribution using the D’Agostino-Pearson omnibus normality test. Comparisons were performed using the Student’s t-test if data were found to have a Gaussian distribution. Alternatively, if data did not have a Gaussian distribution, comparisons were performed using the Mann Whitney test. Data are presented as mean ± SEM. Survival curves were analyzed using the Log-Rank test. All data were analyzed using the statistical software program Prism 4.0 (GraphPad, San Diego, CA), and a p value of <0.05 was considered to be statistically significant.

Results

Mortality is worsened in aged septic mice with impaired lipid transport

To determine the functional significance of impaired lipid transport, aged WT and Mttp-IKO mice were subjected to P. aeruginosa pneumonia and followed 7 days for survival (Figure 1). Septic WT mice had a 39% seven-day mortality, while septic Mttp-IKO mice had an 80% mortality.

thumbnail
Figure 1. Effect of impaired intestinal lipid transport on mortality following P. aeruginosa pneumonia.

Mice with intestine-specific deletion of microsomal triglyceride transfer protein had markedly increased mortality compared to WT mice when followed 7 days after induction of sepsis (n = 18–20/group, p = 0.005).

https://doi.org/10.1371/journal.pone.0101828.g001

Intestinal triglyceride mobilization is impaired in aged septic Mttp-IKO mice

Defects in chylomicron assembly were associated with large intracellular lipid droplets in aged septic Mttp-IKO mice (Fig. 2A, B). Mucosal triglyceride content was increased as expected in aged septic Mttp-IKO mice along with the expected decrease in hepatic triglyceride content as previously observed in young Mttp-IKO mice [60], [65] (Fig. 2C, D). By contrast, mucosal and liver cholesterol content was similar between aged septic Mttp-IKO and WT mice (Fig. 2E, F).

thumbnail
Figure 2. Effect of impaired intestinal lipid transport on intestinal and liver lipids.

Villi of aged septic WT (A) and Mttp-IKO (B) mice stained with osmium tetroxide to detect intracellular lipid droplets. A massive upregulation is noted in these representative histomicrographs in Mttp-IKO mice, (magnification 40x). Triglyceride concentrations were increased in the jejunal mucosa (C, n = 5/group, p = 0.008) but not in the liver (D, n = 5/group, p = 0.056). Cholesterol levels were similar in both jejunal mucosa (E, n = 5/group, p>0.05) and liver (F, n = 5/group, p>0.05).

https://doi.org/10.1371/journal.pone.0101828.g002

Increased Intestinal epithelial apoptosis in aged septic mice with impaired intestinal lipid transport

Intestinal epithelial apoptosis was significantly increased in aged septic Mttp-IKO mice when assayed by H&E or active caspase 3 staining (Fig. 3A, B). The ratio of jejunal Bax/Bcl-2 (Fig. 3C) and Bax/Bcl-xL (Fig. 3D) was also increased in aged septic Mttp-IKO mice compared to aged septic WT mice, consistent with elevated gut apoptosis. These findings are at variance with our prior observations in young septic Mttp-IKO mice [59] and suggest that the intestinal apoptotic response to systemic infection is age-dependent.

thumbnail
Figure 3. Effect of impaired intestinal lipid transport on intestinal epithelial apoptosis.

Intestinal epithelial apoptosis was increased in Mttp-IKO mice by H&E staining (A, n = 11–16/group, p = .002) with a similar trend seen with active caspase 3 staining (B, n = 11–12/group, p = 0.07). The ratio of pro-apoptotic Bax to anti-apoptotic Bcl-2 (C, n = 11–16/group, p = 0.02) and Bcl-xL (D, n = 8–11/group, p = 0.004) were both increased in Mttp-IKO mice, consistent with elevated apoptosis.

https://doi.org/10.1371/journal.pone.0101828.g003

Intestinal length and crypt proliferation are higher in aged septic mice with impaired lipid transport

Villus length was significantly increased in aged Mttp-IKO mice subjected to pneumonia compared to aged WT mice subjected to the same insult (Fig. 4A, B). Crypt depth was also significantly increased in aged septic Mttp-IKO mice (Fig. 4C). This was associated with a significant increase in the number of proliferating S phase crypt cells in aged septic Mttp-IKO mice as measured by BrdU staining (Fig. 4D). These findings recapitulate those in young, septic Mttp-IKO mice [59], suggesting that elements of the response to systemic infection following attenuated chylomicron formation are retained with age.

thumbnail
Figure 4. Effect of impaired intestinal lipid transport on villus and crypt length and proliferation.

Villus length was longer in in Mttp-IKO mice whether assayed qualitatively (A shows representative micrographs, magnification 10X) or quantitatively (B, n = 11–16/group, p<0.0001). Crypt depth was also increased in Mttp-IKO mice (C, n = 11–16/group, p<0.0001). The number of s phase cells (D, n = 11–12/group, p = 0.002) was also significantly higher in Mttp-IKO mice.

https://doi.org/10.1371/journal.pone.0101828.g004

Pulmonary MPO levels are increased in aged septic mice with impaired intestinal lipid transport

Since the infection in pneumonia is initiated in the lungs, pulmonary injury was assayed next. MPO activity was significantly elevated in BAL fluid of aged septic Mttp-IKO mice (Fig, 5A). Despite this, bacteria were undetectable in the majority of both septic WT and Mttp-IKO mice, and bacterial burden was low even in animals from which P. aeruginosa could be recovered (Fig. 5B). Pulmonary cytokines were generally similar between groups except for a decrease in TNFα in aged septic Mttp-IKO mice (Fig, 5C–H).

thumbnail
Figure 5. Effect of impaired intestinal lipid transport on BAL MPO, cultures and cytokines.

MPO levels were elevated in BAL fluid of Mttp-IKO mice (A, n = 8/group, p = 0.001). Bacterial levels were low in both WT and Mttp-IKO mice, although the latter had less bacteria present in BAL fluid (B, n = 9–11/group, p = 0.005). TNFα was lower in BAL fluid in Mttp-IKO mice (C, n = 7–8/group, p = 0.02) but no statistically significant differences were noted in IL-6, MCP-1, IL-1β, IL-10 and IL-13 (D–H, n = 8–11/group, p>0.05 for each).

https://doi.org/10.1371/journal.pone.0101828.g005

Systemic pro-inflammatory cytokines are decreased in aged septic mice with impaired intestinal lipid transport

IL-6, MCP-1 and TNFα levels were all significantly decreased 24 hours after pneumonia in aged septic Mttp-IKO mice (Figure 6A–C). In contrast, there were no significant differences in the serum levels of IL-1β, IL-10 and IL-13 by genotype (Figure 6D–F).

thumbnail
Figure 6. Effect of impaired intestinal lipid transport on systemic cytokines.

Serum TNFα (A, n = 9–12/group, p = 0.0001), IL-6 (B, n = 10–14/group, p = 0.008), MCP-1 (C, n = 10–12/group, p = 0.03) were all lower in Mttp-IKO mice but no statistically significant differences were noted in IL-1β, IL-10 and IL-13 (D–F, n = 10–16/group, p>0.05 for each).

https://doi.org/10.1371/journal.pone.0101828.g006

Liver and splenic blastoid cells are increased in aged septic mice with impaired intestinal lipid transport

The number of liver blastoid cells was increased in aged septic Mttp-IKO mice (Fig. 7A), although there were no significant differences in the frequency of liver hematolymphoid cells or enlarged, regenerative hepatocytes (Fig. 7B). Similarly, the number of splenic blastoid cells was also increased in aged septic Mttp-IKO mice (Fig. 7D). A marked increase in splenic megakaryocytes was also identified (Fig. 7E) although no difference was detected in splenic tingible body macrophages (Fig. 7F).

thumbnail
Figure 7. Effect of impaired intestinal lipid transport on liver and spleen cells.

In the liver, there was a slight increase in blastoid cells (identified by enlargement, hyperchromasia, and pleomorphism) in Mttp-IKO mice (A, n = 10–11/group, p = 0.04) but no difference in hematolymphoid cells (B, n = 10–11/group, p>0.05). In the spleen, there were increases in blastoid cells (C, n = 10–11/group, p = 0.0006) and megakaryocytes (D, n = 10–11/group, p = 0.001) in Mttp-IKO mice but no differences in tingible body macrophages (E, n = N = 10–11/group, p>0.05).

https://doi.org/10.1371/journal.pone.0101828.g007

Impact of sepsis, impaired intestinal lipid transport and combination on TG and cholesterol levels

TG and cholesterol concentrations in serum were both decreased by sepsis in WT mice (Fig. 8A, B). Impaired intestinal lipid transport was verified in aged Mttp-IKO mice with reduced serum TG and cholesterol concentrations. However, while TG levels were similar pre- and post- sepsis in Mttp-IKO mice, serum cholesterol levels rose modestly after the onset of sepsis in Mttp-IKO mice as noted previously in young septic Mttp-IKO mice [59]. Lipoprotein distribution as inferred from FPLC fractionation demonstrated that control septic mice have reduced lipid content in large triglyceride rich lipoproteins (VLDL, fraction 10), while septic Mttp-IKO mice showed a rise in HDL cholesterol, as previously noted in young septic Mttp-IKO mice [59] (Fig. 8C–D). These findings again verify that important elements of the lipid homeostatic responses noted in Mttp-IKO mice are retained in aged mice. Serum bile acids were also examined since previous reports have demonstrated elevated levels in association with liver dysfunction in humans [66]. Bile acid levels were similar between aged septic WT and aged septic Mttp-IKO mice (Fig. 8E).

thumbnail
Figure 8. Effect of impaired intestinal lipid transport on serum lipoproteins.

Triglyceride (A) and cholesterol (B) in serum were measured before and 24 hours after induction of pneumonia in the same mice. Sepsis decreased both triglyceride and cholesterol after induction of pneumonia in WT mice (C, D, n = 16–18/group, p<0.0001 for both). Inhibiting chylomicron assembly also resulted in lower triglyceride and cholesterol at baseline (n = 17–18/group, p<0.0001 for both). In contrast, sepsis did not alter triglyceride levels in Mttp-IKO mice (n = 11–17/group, p>0.05). Additionally, sepsis increased cholesterol levels in Mttp-IKO mice after induction of pneumonia (n = 11–17/group, p<0.0001). Sepsis virtually eliminated triglyceride from large, VLDL and LDL size lipoproteins in WT mice (note fractions 10 and 25, respectively). By contrast, Mttp-IKO mice exhibited virtually no triglyceride in lipoprotein particles, as expected. Sepsis resulted in an increase in HDL cholesterol content in Mttp-IKO mice (fractions 38–40). Serum bile acids (E) were similar between WT and Mttp-IKO mice (n = 7–8/group, p>0.05).

https://doi.org/10.1371/journal.pone.0101828.g008

Discussion

The central finding of this study is that mortality is markedly increased in aged septic Mttp-IKO mice following P. aeruginosa-induced sepsis. This observation directly contradicts findings in genetically identical young mice where blocking intestinal chylomicron assembly results in a survival benefit. Since the major difference in experimental design between this study and our prior study on young septic Mttp-IKO mice [59] is mouse age, it is reasonable to conclude that age plays an important role in the divergent survival in young vs. aged Mttp-IKO mice subjected to sepsis. This is consistent with multiple previous studies that have demonstrated that age greatly impacts the host response in sepsis, and interventions that are beneficial in young septic hosts fail to improve or even worsen mortality in aged hosts [4][12].

When trying to understand the differential mortality between young and aged septic Mttp-IKO mice, it is helpful to compare some of the key physiologic and cellular parameters that we observed. Parameters that are similarly changed between young and aged Mttp-IKO mice compared to WT animals – such as villus length, crypt proliferation, pulmonary and systemic cytokines – are unlikely to play a causative role in the differential mortality observed. In contrast, parameters in which results move in the opposite direction (i.e. increased vs. decreased) or change in only one group (i.e. increased vs. no difference) between young and aged Mttp-IKO mice animals compared to WT animals could potentially play a role in mediating the differential mortality observed. It is notable, then, that of the multiple parameters examined in this study, only gut apoptosis, pulmonary neutrophil infiltration, systemic TNFΑ, and mucosal triglyceride concentration were different in young and aged septic Mttp-IKO mice when compared to WT mice.

Gut epithelial apoptosis is increased in both mouse models and human autopsy studies of sepsis [20], [21], [67]. This appears to be physiologically significant since overexpression of Bcl-2 is associated with increased survival from sepsis from either P. aeruginosa or cecal ligation and puncture (CLP) [20], [21]. Numerous co-morbidities including cancer and chronic alcohol abuse are associated with increased sepsis-induced gut epithelial apoptosis. Notably aging is also associated with increased gut apoptosis [29], [33], [34]. Gut apoptosis is increased in aged Mttp-IKO mice in this study, associated with increased ratios of both jejunal Bax/Bcl-2 and Bax/Bcl-xL. Sepsis in isolation from either methicillin resistant Staphylococcus aureus pneumonia or peritonitis increase both pro (Bax) and anti (Bcl-xL) apoptotic mediators in the mitochondrial pathway [28], [30], and the ratio of these plays a critical role in determining whether a cell lives or dies. The finding that gut apoptosis is increased in aged septic Mttp-IKO mice directly contrasts with young septic Mttp-IKO mice, where gut apoptosis was reduced. However, the mechanisms through which blocking intestinal chylomicron alter gut apoptosis are unclear. Further, the association between gut apoptosis and mortality does not distinguish whether gut apoptosis is a mediator of mortality in these studies or simply a marker of illness.

An association also exists between pulmonary neutrophil infiltration and mortality in Mttp-IKO mice, with increased MPO activity and mortality in aged septic mice while MPO activity was unchanged in young septic Mttp-IKO mice compared to aged matched septic WT mice [59]. Pulmonary neutrophil infiltration is commonly seen in the acute respiratory distress syndrome [68][70]. While neutrophils play a beneficial role against pulmonary pathogens, they also release toxic factors such as reactive oxygen species and proteinases that contribute to progression of lung disease. We do acknowledge that is counterintuitive that mice with lower levels of pulmonary bacteria have a higher mortality. However, most mice (both WT and knockout) had no bacteria recovered from the lung 24 hours after the onset of pneumonia and the average bacterial burden was only 100 cfu/ml in WT mice. In the absence of detectable infection in most mice, it is unlikely that the small difference in pulmonary bacterial burden played a major role in mediating higher mortality in aged septic Mttp-IKO mice. However, excessive pulmonary inflammation, independent of infection, can lead to self-sustaining damage to lung tissue and then secondarily lead to worsening systemic illness. As such, increased pulmonary neutrophil infiltration represents a plausible potential mechanism underlying mortality in aged septic Mttp-IKO mice. Of note, in hemorrhagic shock, toxic mediators are released from the intestine and transported through the mesenteric lymph to the pulmonary circulation. Studies in rats have demonstrated that mesenteric lymph duct ligation prevents lung injury and improves survival in multiple animal models of critical illness [19], [50], [52], [71], [72]. The lungs do not appear to be a major contributor towards improved survival in young septic Mttp-IKO mice, mice since decrease of lipid transport into mesenteric lymph did not prevent lung injury. Accordingly, further work will be required to resolve the mechanisms through which blocking intestinal chylomicron alters pulmonary inflammation in an age-dependent manner after sepsis.

One surprising finding was that Mttp-IKO mice generally had lower levels of systemic cytokines. Typically, this would be thought of as being associated with better outcomes rather than higher mortality. However, it should be noted that a similar pattern of lower systemic cytokines was seen in both aged and young Mttp-IKO mice compared to age-matched WT mice. This suggests that cytokine levels alone cannot be responsible for the differential mortality seen between aged and young mice. It is unclear, however, whether systemic cytokines played no role in survival or if a beneficial role was superseded by a different detrimental mechanism in aged mice. In this regard, it should be mentioned that bacteremia was not different between aged Mttp-IKO and WT mice (data not shown).

Patients in the ICU frequently have decreased serum lipids [73]. The functional significance of this observation is not clear. In preclinical models, lipoprotein infusion improves mortality in endotoxemia [74], a model of critical illness with some similarities but also some key differences from sepsis. Alternatively, hyperlipoproteinemic LDL receptor-deficient mice have increased mortality following CLP [75]. It is noteworthy that the adaptive homeostatic responses to sepsis noted in young Mttp-IKO mice (increased HDL cholesterol content) appear to be recapitulated in aged mice, but again it is unlikely that this is a determinant of the paradoxical increase noted in mortality.

Another, albeit less likely, factor that could potentially play a role in the differential mortality between young and aged mice is severity of insult. Given that aged mice are more susceptible to sepsis, it is necessary to lessen insult severity in aged animals in order to match mortality between young and aged animals, since aged animals have 100% mortality to sepsis models that cause moderate lethality in younger mice [4]. Both age and severity of insult independently mediate survival and alter the host response following CLP, so it is possible that a differential bacterial burden (rather than age) played a critical role in mediating mortality in this study. Additionally, as alluded to above, while we attempted to match mortality between young and aged mice, baseline mortality from P. aeruginosa was 29% in our previous study of young mice whereas baseline mortality from the same organism was 39% in this study. While this difference is relatively small, the risk of death prior to intervention has been shown to play a large role in how (or if) a host responds to a putative sepsis therapeutic [76], [77], and it is possible that this seemingly small difference in baseline mortality was associated with a host milieu that was sufficiently different that blocking intestinal chylomicrons had a profoundly different effect.

This study has a number of limitations. All mechanistic studies were performed at 24 hours, and we therefore do not know how or if any of the parameters altered in aged septic Mttp-IKO mice changed over time compared to septic WT mice. This especially limits interpretation of the cytokine data, in light of the fact that cytokine levels are highly dynamic following sepsis [78], [79]. Further, although we compared septic aged matched Mttp-IKO and WT mice, it is important to recognize that intestinal Mttp deletion was imposed for only 3–4 weeks in these mice. The effects of sustained Mttp deletion in aging mice was not examined and it is unknown whether these mice would tolerate virtually lifelong attenuation of lipid absorption. Accordingly, while we previously compared young unmanipulated Mttp-IKO and WT mice, it is possible that aging results in local and systemic phenotypic differences between these animals not seen in young mice. As such, some of the differences noted in aged septic Mttp-IKO and WT mice might have already been different prior to the onset of pneumonia. It is also important to mention that the mice used were generated from two inbred strains. There is significant controversy as to whether inbred animals are appropriate surrogates for humans which obviously have more genetic diversity [14], [80], [81], and our results must be interpreted in light of the fact that they were derived from two inbred strains. In addition, excision and homogenization of the entire lung would likely have provided a more accurate assessment of pulmonary endpoints examined. Finally, we acknowledge that the associations between increased mortality and the physiologic parameters measured do not imply causation, and additional studies are needed to determine the significance of differences identified between septic aged matched Mttp-IKO and WT mice.

Despite these limitations, this study highlights the importance of age in the pathophysiology of sepsis. Further, it demonstrates that blocking intestinal chylomicron secretion can have widely varying results in sepsis, which must be taken into account when considering the role of mesenteric lymph in sepsis. Further research is needed to identify the mechanisms through which blocking intestinal chylomicron assembly alters outcomes in sepsis.

Author Contributions

Conceived and designed the experiments: CC ND JD. Performed the experiments: ZL YX E. Breed BY E. Burd AF. Analyzed the data: ZL YX E. Burd AF ND CC. Contributed reagents/materials/analysis tools: E. Burd ND CC. Contributed to the writing of the manuscript: ZL YX JD BY E. Burd AF ND CC.

References

  1. 1. Gaieski DF, Edwards JM, Kallan MJ, Carr BG (2013) Benchmarking the incidence and mortality of severe sepsis in the United States. Crit Care Med 41: 1167–1174 10.1097/CCM.0b013e31827c09f8 [doi].
  2. 2. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, et al. (2001) Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 29: 1303–1310.
  3. 3. Martin GS, Mannino DM, Moss M (2006) The effect of age on the development and outcome of adult sepsis. Crit Care Med 34: 15–21.
  4. 4. Turnbull IR, Clark AT, Stromberg PE, Dixon DJ, Woolsey CA, et al. (2009) Effects of aging on the immunopathologic response to sepsis. Crit Care Med 37: 1018–1023.
  5. 5. Tucsek Z, Gautam T, Sonntag WE, Toth P, Saito H, et al. (2013) Aging exacerbates microvascular endothelial damage induced by circulating factors present in the serum of septic patients. J Gerontol A Biol Sci Med Sci 68: 652–660. gls232 [pii]; 10.1093/gerona/gls232 [doi].
  6. 6. Drechsler S, Weixelbaumer K, Raeven P, Jafarmadar M, Khadem A, et al. (2012) Relationship between age/gender-induced survival changes and the magnitude of inflammatory activation and organ dysfunction in post-traumatic sepsis. PLoS ONE 7: e51457 10.1371/journal.pone.0051457 [doi];PONE-D-12-24896 [pii].
  7. 7. Cao Z, Yende S, Kellum JA, Angus DC, Robinson RA (2013) Proteomics Reveals Age-related Differences in the Host Immune Response to Sepsis. J Proteome Res. 10.1021/pr400814s [doi].
  8. 8. Opal SM, Girard TD, Ely EW (2005) The immunopathogenesis of sepsis in elderly patients. Clin Infect Dis 41 Suppl 7S504–S512.
  9. 9. Yamamoto K, Shimokawa T, Yi H, Isobe K, Kojima T, et al. (2002) Aging accelerates endotoxin-induced thrombosis: increased responses of plasminogen activator inhibitor-1 and lipopolysaccharide signaling with aging. Am J Pathol 161: 1805–1814. S0002-9440(10)64457-4 [pii].
  10. 10. Saito H, Sherwood ER, Varma TK, Evers BM (2003) Effects of aging on mortality, hypothermia, and cytokine induction in mice with endotoxemia or sepsis. Mech Ageing Dev 124: 1047–1058.
  11. 11. Gomez CR, Karavitis J, Palmer JL, Faunce DE, Ramirez L, et al. (2010) Interleukin-6 contributes to age-related alteration of cytokine production by macrophages. Mediators Inflamm 2010: 475139 10.1155/2010/475139 [doi].
  12. 12. Chen MM, Palmer JL, Plackett TP, Deburghgraeve CR, Kovacs EJ (2014) Age-related differences in the neutrophil response to pulmonary pseudomonas infection. Exp Gerontol. S0531-5565(13)00366-5 [pii]; 10.1016/j.exger.2013.12.010 [doi].
  13. 13. Turnbull IR, Wlzorek JJ, Osborne D, Hotchkiss RS, Coopersmith CM, et al. (2003) Effects of age on mortality and antibiotic efficacy in cecal ligation and puncture. Shock 19: 310–313.
  14. 14. Fink MP (2014) Animal models of sepsis. Virulence 5: 143–153. 26083 [pii]; 10.4161/viru.26083 [doi].
  15. 15. Ward PA (2012) New approaches to the study of sepsis. EMBO Mol Med 4: 1234–1243 10.1002/emmm.201201375 [doi].
  16. 16. Mittal R, Coopersmith CM (2013) Redefining the gut as the motor of critical illness. Trends Mol Med. S1471-4914(13)00147-0 [pii]; 10.1016/j.molmed.2013.08.004 [doi].
  17. 17. Clark JA, Coopersmith CM (2007) Intestinal crosstalk: a new paradigm for understanding the gut as the “motor” of critical illness. Shock 28: 384–393.
  18. 18. Carrico CJ, Meakins JL, Marshall JC, Fry D, Maier RV (1986) Multiple-organ-failure syndrome. The gastrointestinal tract: the “motor” of MOF. Arch Surg 121: 196–208.
  19. 19. Deitch EA (2012) Gut-origin sepsis: evolution of a concept. Surgeon 10: 350–356. S1479-666X(12)00033-9 [pii]; 10.1016/j.surge.2012.03.003 [doi].
  20. 20. Coopersmith CM, Stromberg PE, Dunne WM, Davis CG, Amiot DM, et al. (2002) Inhibition of intestinal epithelial apoptosis and survival in a murine model of pneumonia-induced sepsis. JAMA 287: 1716–1721.
  21. 21. Coopersmith CM, Chang KC, Swanson PE, Tinsley KW, Stromberg PE, et al. (2002) Overexpression of Bcl-2 in the intestinal epithelium improves survival in septic mice. Crit Care Med 30: 195–201.
  22. 22. Coopersmith CM, Stromberg PE, Davis CG, Dunne WM, Amiot DM, et al. (2003) Sepsis from Pseudomonas aeruginosa pneumonia decreases intestinal proliferation and induces gut epithelial cell cycle arrest. Crit Care Med 31: 1630–1637.
  23. 23. Clark JA, Clark AT, Hotchkiss RS, Buchman TG, Coopersmith CM (2008) Epidermal growth factor treatment decreases mortality and is associated with improved gut integrity in sepsis. Shock 30: 36–42.
  24. 24. Clark JA, Gan H, Samocha AJ, Fox AC, Buchman TG, et al. (2009) Enterocyte-specific epidermal growth factor prevents barrier dysfunction and improves mortality in murine peritonitis. Am J Physiol Gastrointest Liver Physiol 297: G471–G479.
  25. 25. Dominguez JA, Coopersmith CM (2010) Can we protect the gut in critical illness? The role of growth factors and other novel approaches. Crit Care Clin 26: 549–65, x. S0749-0704(10)00025-4 [pii]; 10.1016/j.ccc.2010.04.005 [doi].
  26. 26. Dominguez JA, Samocha AJ, Liang Z, Burd EM, Farris AB, et al. (2013) Inhibition of IKKbeta in Enterocytes Exacerbates Sepsis-Induced Intestinal Injury and Worsens Mortality. Crit Care Med 41: e275–e285 10.1097/CCM.0b013e31828a44ed [doi].
  27. 27. Fox AC, McConnell KW, Yoseph BP, Breed E, Liang Z, et al. (2012) The endogenous bacteria alter gut epithelial apoptosis and decrease mortality following Pseudomonas aeruginosa pneumonia. Shock 38: 508–514 10.1097/SHK.0b013e31826e47e8 [doi].
  28. 28. Stromberg PE, Woolsey CA, Clark AT, Clark JA, Turnbull IR, et al. (2009) CD4+ lymphocytes control gut epithelial apoptosis and mediate survival in sepsis. FASEB J 23: 1817–1825.
  29. 29. Yoseph BP, Breed E, Overgaard CE, Ward CJ, Liang Z, et al. (2013) Chronic alcohol ingestion increases mortality and organ injury in a murine model of septic peritonitis. PLoS ONE 8: e62792 10.1371/journal.pone.0062792 [doi];PONE-D-13-07625 [pii].
  30. 30. Perrone EE, Jung E, Breed E, Dominguez JA, Liang Z, et al. (2012) Mechanisms of methicillin-resistant Staphylococcus aureus pneumonia-induced intestinal epithelial apoptosis. Shock 38: 68–75 10.1097/SHK.0b013e318259abdb [doi].
  31. 31. Rupani B, Caputo FJ, Watkins AC, Vega D, Magnotti LJ, et al. (2007) Relationship between disruption of the unstirred mucus layer and intestinal restitution in loss of gut barrier function after trauma hemorrhagic shock. Surgery 141: 481–489. S0039-6060(06)00710-0 [pii]; 10.1016/j.surg.2006.10.008 [doi].
  32. 32. Fink MP (2003) Intestinal epithelial hyperpermeability: update on the pathogenesis of gut mucosal barrier dysfunction in critical illness. Curr Opin Crit Care 9: 143–151.
  33. 33. Fox AC, Robertson CM, Belt B, Clark AT, Chang KC, et al. (2010) Cancer causes increased mortality and is associated with altered apoptosis in murine sepsis. Crit Care Med 38: 886–893 10.1097/CCM.0b013e3181c8fdb1 [doi].
  34. 34. Turnbull IR, Buchman TG, Javadi P, Woolsey CA, Hotchkiss RS, et al. (2004) Age disproportionately increases sepsis-induced apoptosis in the spleen and gut epithelium. Shock 22: 364–368.
  35. 35. Vyas D, Robertson CM, Stromberg PE, Martin JR, Dunne WM, et al. (2007) Epithelial apoptosis in mechanistically distinct methods of injury in the murine small intestine. Histol Histopathol 22: 623–630.
  36. 36. Chang M, Alsaigh T, Kistler EB, Schmid-Schonbein GW (2012) Breakdown of mucin as barrier to digestive enzymes in the ischemic rat small intestine. PLoS ONE 7: e40087 10.1371/journal.pone.0040087 [doi];PONE-D-12-08417 [pii].
  37. 37. Chang M, Kistler EB, Schmid-Schonbein GW (2012) Disruption of the mucosal barrier during gut ischemia allows entry of digestive enzymes into the intestinal wall. Shock 37: 297–305 10.1097/SHK.0b013e318240b59b [doi].
  38. 38. Qin X, Caputo FJ, Xu DZ, Deitch EA (2008) Hydrophobicity of mucosal surface and its relationship to gut barrier function. Shock 29: 372–376 10.1097/shk.0b013e3181453f4e [doi]; 00024382-200803000-00010 [pii].
  39. 39. Lu Q, Xu DZ, Sharpe S, Doucet D, Pisarenko V, et al. (2011) The anatomic sites of disruption of the mucus layer directly correlate with areas of trauma/hemorrhagic shock-induced gut injury. J Trauma 70: 630–635 10.1097/TA.0b013e3181e1221b [doi].
  40. 40. Fishman JE, Levy G, Alli V, Sheth S, Lu Q, et al. (2013) Oxidative modification of the intestinal mucus layer is a critical but unrecognized component of trauma hemorrhagic shock-induced gut barrier failure. Am J Physiol Gastrointest Liver Physiol 304: G57–G63. ajpgi.00170.2012 [pii]; 10.1152/ajpgi.00170.2012 [doi].
  41. 41. Chen C, Wang P, Su Q, Wang S, Wang F (2012) Myosin light chain kinase mediates intestinal barrier disruption following burn injury. PLoS ONE 7: e34946 10.1371/journal.pone.0034946 [doi];PONE-D-11-24867 [pii].
  42. 42. Turner JR (2009) Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 9: 799–809. nri2653 [pii]; 10.1038/nri2653 [doi].
  43. 43. Geng Y, Li J, Wang F, Li Q, Wang X, Sun L, et al. (2013) Epidermal growth factor promotes proliferation and improves restoration after intestinal ischemia-reperfusion injury in rats. Inflammation 36: 670–679 10.1007/s10753-012-9591-x [doi].
  44. 44. Yang J, Radulescu A, Chen CL, Zhang HY, James IO, et al. (2013) Heparin-binding epidermal growth factor-like growth factor improves intestinal barrier function and reduces mortality in a murine model of peritonitis. Surgery 153: 52–62. S0039-6060(12)00168-7 [pii]; 10.1016/j.surg.2012.04.002 [doi].
  45. 45. Seal JB, Alverdy JC, Zaborina O, An G (2011) Agent-based dynamic knowledge representation of Pseudomonas aeruginosa virulence activation in the stressed gut: Towards characterizing host-pathogen interactions in gut-derived sepsis. Theor Biol Med Model 8: 33. 1742-4682-8-33 [pii]; 10.1186/1742-4682-8-33 [doi].
  46. 46. Fink D, Romanowski K, Valuckaite V, Babrowski T, Kim M, et al. (2011) Pseudomonas aeruginosa potentiates the lethal effect of intestinal ischemia-reperfusion injury: the role of in vivo virulence activation. J Trauma 71: 1575–1582 10.1097/TA.0b013e31821cb7e5 [doi].
  47. 47. Babrowski T, Romanowski K, Fink D, Kim M, Gopalakrishnan V, et al. (2013) The intestinal environment of surgical injury transforms Pseudomonas aeruginosa into a discrete hypervirulent morphotype capable of causing lethal peritonitis. Surgery 153: 36–43. S0039-6060(12)00315-7 [pii]; 10.1016/j.surg.2012.06.022 [doi].
  48. 48. Romanowski K, Zaborin A, Valuckaite V, Rolfes RJ, Babrowski T, et al. (2012) Candida albicans isolates from the gut of critically ill patients respond to phosphate limitation by expressing filaments and a lethal phenotype. PLoS ONE 7: e30119 10.1371/journal.pone.0030119 [doi];PONE-D-11-15176 [pii].
  49. 49. Deitch EA (2010) Gut lymph and lymphatics: a source of factors leading to organ injury and dysfunction. Ann N Y Acad Sci 1207 Suppl 1E103–E111 10.1111/j.1749-6632.2010.05713.x [doi].
  50. 50. Watkins AC, Caputo FJ, Badami C, Barlos D, Xu dZ, et al. (2008) Mesenteric lymph duct ligation attenuates lung injury and neutrophil activation after intraperitoneal injection of endotoxin in rats. J Trauma 64: 126–130 10.1097/TA.0b013e3181574a8a [doi]; 00005373-200801000-00020 [pii].
  51. 51. Sambol JT, Lee MA, Caputo FJ, Kawai K, Badami C, et al. (2009) Mesenteric lymph duct ligation prevents trauma/hemorrhage shock-induced cardiac contractile dysfunction. J Appl Physiol 106: 57–65. 90937.2008 [pii]; 10.1152/japplphysiol.90937.2008 [doi].
  52. 52. Fang JF, Shih LY, Yuan KC, Fang KY, Hwang TL, et al. (2010) Proteomic analysis of post-hemorrhagic shock mesenteric lymph. Shock 34: 291–298 10.1097/SHK.0b013e3181ceef5e [doi].
  53. 53. Mittal A, Middleditch M, Ruggiero K, Loveday B, Delahunt B, et al. (2010) Changes in the mesenteric lymph proteome induced by hemorrhagic shock. Shock 34: 140–149 10.1097/SHK.0b013e3181cd8631 [doi].
  54. 54. Peltz ED, Moore EE, Zurawel AA, Jordan JR, Damle SS, et al. (2009) Proteome and system ontology of hemorrhagic shock: exploring early constitutive changes in postshock mesenteric lymph. Surgery 146: 347–357. S0039-6060(09)00260-8 [pii]; 10.1016/j.surg.2009.02.022 [doi].
  55. 55. Deitch EA, Qin X, Sheth SU, Tiesi G, Palange D, et al. (2011) Anticoagulants influence the in vitro activity and composition of shock lymph but not its in vivo activity. Shock 36: 177–183 10.1097/SHK.0b013e3182205c30 [doi].
  56. 56. Reino DC, Pisarenko V, Palange D, Doucet D, Bonitz RP, et al. (2011) Trauma hemorrhagic shock-induced lung injury involves a gut-lymph-induced TLR4 pathway in mice. PLoS ONE 6: e14829 10.1371/journal.pone.0014829 [doi]; 09-PONE-RA-14427R1 [pii].
  57. 57. Reino DC, Palange D, Feketeova E, Bonitz RP, Xu dZ, et al. (2012) Activation of toll-like receptor 4 is necessary for trauma hemorrhagic shock-induced gut injury and polymorphonuclear neutrophil priming. Shock 38: 107–114 10.1097/SHK.0b013e318257123a [doi].
  58. 58. Berriot-Varoqueaux N, Aggerbeck LP, Samson-Bouma M, Wetterau JR (2000) The role of the microsomal triglygeride transfer protein in abetalipoproteinemia. Annu Rev Nutr 20: 663–697.
  59. 59. Dominguez JA, Xie Y, Dunne WM, Yoseph BP, Burd EM, et al. (2012) Intestine-specific Mttp deletion decreases mortality and prevents sepsis-induced intestinal injury in a murine model of Pseudomonas aeruginosa pneumonia. PLoS ONE 7: e49159 10.1371/journal.pone.0049159 [doi];PONE-D-12-07223 [pii].
  60. 60. Xie Y, Newberry EP, Young SG, Robine S, Hamilton RL, et al. (2006) Compensatory increase in hepatic lipogenesis in mice with conditional intestine-specific Mttp deficiency. J Biol Chem 281: 4075–4086.
  61. 61. Nemzek JA, Xiao HY, Minard AE, Bolgos GL, Remick DG (2004) Humane endpoints in shock research. Shock 21: 17–25.
  62. 62. Nemzek JA, Hugunin KM, Opp MR (2008) Modeling sepsis in the laboratory: merging sound science with animal well-being. Comp Med 58: 120–128.
  63. 63. McConnell KW, McDunn JE, Clark AT, Dunne WM, Dixon DJ, et al. (2010) Streptococcus pneumoniae and Pseudomonas aeruginosa pneumonia induce distinct host responses. Crit Care Med 38: 223–241 10.1097/CCM.0b013e3181b4a76b [doi].
  64. 64. Dominguez JA, Vithayathil PJ, Khailova L, Lawrance CP, Samocha AJ, et al. (2011) Epidermal growth factor improves survival and prevents intestinal injury in a murine model of pseudomonas aeruginosa pneumonia. Shock 36: 381–389 10.1097/SHK.0b013e31822793c4 [doi].
  65. 65. Xie Y, Kennedy S, Sidhu R, Luo J, Ory DS, et al. (2012) Liver X receptor agonist modulation of cholesterol efflux in mice with intestine-specific deletion of microsomal triglyceride transfer protein. Arterioscler Thromb Vasc Biol 32: 1624–1631. ATVBAHA.112.246066 [pii]; 10.1161/ATVBAHA.112.246066 [doi].
  66. 66. Muraca M, Kohlhaw K, Vilei MT, Ringe B, Bunzendahl H, et al. (1993) Serum bile acids and esterified bilirubin in early detection and differential diagnosis of hepatic dysfunction following orthotopic liver transplantation. J Hepatol 17: 141–145.
  67. 67. Hiramatsu M, Hotchkiss RS, Karl IE, Buchman TG (1997) Cecal ligation and puncture (CLP) induces apoptosis in thymus, spleen, lung, and gut by an endotoxin and TNF-independent pathway. Shock 7: 247–253.
  68. 68. Williams AE, Chambers RC (2014) The mercurial nature of neutrophils: still an enigma in ARDS? Am J Physiol Lung Cell Mol Physiol 306: L217–L230. ajplung.00311.2013 [pii]; 10.1152/ajplung.00311.2013 [doi]
  69. 69. Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, et al. (2012) Acute respiratory distress syndrome: the Berlin Definition. JAMA 307: 2526–2533. 1160659 [pii]; 10.1001/jama.2012.5669 [doi].
  70. 70. Zhou X, Dai Q, Huang X (2012) Neutrophils in acute lung injury. Front Biosci (Landmark Ed) 17: 2278–2283. 4051 [pii].
  71. 71. Badami CD, Senthil M, Caputo FJ, Rupani BJ, Doucet D, et al. (2008) Mesenteric lymph duct ligation improves survival in a lethal shock model. Shock 30: 680–685 10.1097/SHK.0b013e318173edd1 [doi].
  72. 72. Sarin EL, Moore EE, Moore JB, Masuno T, Moore JL, et al. (2004) Systemic neutrophil priming by lipid mediators in post-shock mesenteric lymph exists across species. J Trauma 57: 950-954. 00005373-200411000-00005 [pii].
  73. 73. Ott J, Hiesgen C, Mayer K (2011) Lipids in critical care medicine. Prostaglandins Leukot Essent Fatty Acids 85: 267–273. S0952-3278(11)00044-5 [pii]; 10.1016/j.plefa.2011.04.011 [doi].
  74. 74. Wu A, Hinds CJ, Thiemermann C (2004) High-density lipoproteins in sepsis and septic shock: metabolism, actions, and therapeutic applications. Shock 21: 210–221.
  75. 75. Lanza-Jacoby S, Miller S, Jacob S, Heumann D, Minchenko AG, et al. (2003) Hyperlipoproteinemic low-density lipoprotein receptor-deficient mice are more susceptible to sepsis than corresponding wild-type mice. J Endotoxin Res 9: 341–347.
  76. 76. Qiu P, Cui X, Barochia A, Li Y, Natanson C, et al. (2011) The evolving experience with therapeutic TNF inhibition in sepsis: considering the potential influence of risk of death. Expert Opin Investig Drugs 20: 1555–1564 10.1517/13543784.2011.623125 [doi].
  77. 77. Eichacker PQ, Parent C, Kalil A, Esposito C, Cui X, et al. (2002) Risk and the efficacy of antiinflammatory agents: retrospective and confirmatory studies of sepsis. Am J Respir Crit Care Med 166: 1197–1205.
  78. 78. Osuchowski MF, Craciun F, Weixelbaumer KM, Duffy ER, Remick DG (2012) Sepsis chronically in MARS: systemic cytokine responses are always mixed regardless of the outcome, magnitude, or phase of sepsis. J Immunol 189: 4648–4656. jimmunol.1201806 [pii]; 10.4049/jimmunol.1201806 [doi].
  79. 79. Osuchowski MF, Welch K, Siddiqui J, Remick DG (2006) Circulating cytokine/inhibitor profiles reshape the understanding of the SIRS/CARS continuum in sepsis and predict mortality. J Immunol 177: 1967–1974.
  80. 80. Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, et al. (2013) Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A 110: 3507–3512. 1222878110 [pii]; 10.1073/pnas.1222878110 [doi].
  81. 81. Osuchowski MF, Remick DG, Lederer JA, Lang CH, Aasen AO, et al. (2014) Abandon the mouse research ship? Not just yet! Shock 41: 463–475 10.1097/SHK.0000000000000153 [doi].