Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Shock ; 46(1): 52-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27299587

ABSTRACT

Intestinal barrier dysfunction is thought to contribute to the development of multiple organ dysfunction syndrome in sepsis. Although there are similarities in clinical course following sepsis, there are significant differences in the host response depending on the initiating organism and time course of the disease, and pathways of gut injury vary widely in different preclinical models of sepsis. The purpose of this study was to determine whether the timecourse and mechanisms of intestinal barrier dysfunction are similar in disparate mouse models of sepsis with similar mortalities. FVB/N mice were randomized to receive cecal ligation and puncture (CLP) or sham laparotomy, and permeability was measured to fluoresceinisothiocyanate conjugated-dextran (FD-4) six to 48 h later. Intestinal permeability was elevated following CLP at all timepoints measured, peaking at 6 to 12 h. Tight junction proteins claudin 1, 2, 3, 4, 5, 7, 8, 13, and 15, Junctional Adhesion Molecule-A (JAM-A), occludin, and ZO-1 were than assayed by Western blot, real-time polymerase chain reaction, and immunohistochemistry 12 h after CLP to determine potential mechanisms underlying increases in intestinal permeability. Claudin 2 and JAM-A were increased by sepsis, whereas claudin-5 and occludin were decreased by sepsis. All other tight junction proteins were unchanged. A further timecourse experiment demonstrated that alterations in claudin-2 and occludin were detectable as early as 1 h after the onset of sepsis. Similar experiments were then performed in a different group of mice subjected to Pseudomonas aeruginosa pneumonia. Mice with pneumonia had an increase in intestinal permeability similar in timecourse and magnitude to that seen in CLP. Similar changes in tight junction proteins were seen in both models of sepsis although mice subjected to pneumonia also had a marked decrease in ZO-1 not seen in CLP. These results indicate that two disparate, clinically relevant models of sepsis induce a significant increase in intestinal permeability mediated through a common pathway involving alterations in claudin 2, claudin 5, JAM-A, and occludin although model-specific differences in ZO-1 were also identified.


Subject(s)
Intestinal Diseases/metabolism , Intestinal Perforation/metabolism , Sepsis/metabolism , Animals , Cecum/injuries , Claudins/genetics , Claudins/metabolism , Female , Intestinal Diseases/pathology , Ligation/adverse effects , Male , Mice , Occludin/genetics , Occludin/metabolism , Pneumonia/genetics , Pneumonia/metabolism , Pseudomonas Infections/genetics , Pseudomonas Infections/metabolism , Sepsis/pathology , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
2.
PLoS One ; 9(7): e101828, 2014.
Article in English | MEDLINE | ID: mdl-25010671

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.


Subject(s)
Aging/metabolism , Carrier Proteins/genetics , Intestinal Mucosa/metabolism , Sepsis/mortality , Sepsis/physiopathology , Aging/genetics , Animals , Apoptosis , Biological Transport , Cell Proliferation , Cholesterol/metabolism , Cytokines/metabolism , Gene Knockout Techniques , Intestinal Mucosa/pathology , Intestines/pathology , Liver/immunology , Lung/metabolism , Mice , Organ Specificity , Peroxidase/metabolism , Pseudomonas aeruginosa/physiology , Sepsis/metabolism , Sepsis/pathology , Spleen/immunology , Survival Analysis , Triglycerides/metabolism
3.
PLoS One ; 8(10): e77203, 2013.
Article in English | MEDLINE | ID: mdl-24204769

ABSTRACT

World conditions place large populations at risk from ionizing radiation (IR) from detonation of dirty bombs or nuclear devices. In a subgroup of patients, ionizing radiation exposure would be followed by a secondary infection. The effects of radiation combined injury are potentially more lethal than either insult in isolation. The purpose of this study was to determine mechanisms of mortality and possible therapeutic targets in radiation combined injury. Mice were exposed to IR with 2.5 Gray (Gy) followed four days later by intratracheal methicillin-resistant Staphylococcus aureus (MRSA). While either IR or MRSA alone yielded 100% survival, animals with radiation combined injury had 53% survival (p = 0.01). Compared to IR or MRSA alone, mice with radiation combined injury had increased gut apoptosis, local and systemic bacterial burden, decreased splenic CD4 T cells, CD8 T cells, B cells, NK cells, and dendritic cells, and increased BAL and systemic IL-6 and G-CSF. In contrast, radiation combined injury did not alter lymphocyte apoptosis, pulmonary injury, or intestinal proliferation compared to IR or MRSA alone. In light of the synergistic increase in gut apoptosis following radiation combined injury, transgenic mice that overexpress Bcl-2 in their intestine and wild type mice were subjected to IR followed by MRSA. Bcl-2 mice had decreased gut apoptosis and improved survival compared to WT mice (92% vs. 42%; p<0.01). These data demonstrate that radiation combined injury results in significantly higher mortality than could be predicted based upon either IR or MRSA infection alone, and that preventing gut apoptosis may be a potential therapeutic target.


Subject(s)
Apoptosis/immunology , Intestinal Mucosa/immunology , Radiation Injuries, Experimental/immunology , Staphylococcal Infections/immunology , Animals , Apoptosis/genetics , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , B-Lymphocytes/radiation effects , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/radiation effects , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/radiation effects , Gamma Rays , Gene Expression , Intestinal Mucosa/microbiology , Intestinal Mucosa/radiation effects , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/radiation effects , Lung/immunology , Lung/microbiology , Lung/radiation effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Radiation Injuries, Experimental/complications , Radiation Injuries, Experimental/mortality , Radiation Injuries, Experimental/pathology , Staphylococcal Infections/complications , Staphylococcal Infections/mortality , Staphylococcal Infections/pathology , Survival Analysis , Whole-Body Counting
4.
Crit Care Med ; 41(10): e275-85, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939348

ABSTRACT

OBJECTIVES: Nuclear factor-κB is a critical regulator of cell-survival genes and the host inflammatory response. The purpose of this study was to investigate the role of enterocyte-specific NF-kB in sepsis through selective ablation of IkB kinase. DESIGN: Prospective, randomized controlled study. SETTING: Animal laboratories in university medical centers. SUBJECTS AND INTERVENTIONS: Mice lacking functional NF-kB in their intestinal epithelium (Vil-Cre/Ikkß) and wild-type mice were subjected to sham laparotomy or cecal ligation and puncture. Animals were killed at 24 hours or followed 7 days for survival. MEASUREMENTS AND MAIN RESULTS: Septic wild-type mice had decreased villus length compared with sham mice, whereas villus atrophy was further exacerbated in septic Vil-Cre/Ikkß mice. Sepsis induced an increase in intestinal epithelial apoptosis compared with sham mice, which was further exacerbated in Vil-Cre/Ikkß mice. Sepsis induced intestinal hyperpermeability in wild-type mice compared with sham mice, which was further exacerbated in septic Vil-Cre/Ikkß mice. This was associated with increased intestinal expression of claudin-2 in septic wild-type mice, which was further increased in septic Vil-Cre/Ikkß mice. Both, pro-inflammatory and anti-inflammatory cytokines were increased in serum following cecal ligation and puncture, and interleukin 10 and monocyte chemoattractant protein-1 levels were higher in septic Vil-Cre/Ikkß mice than in septic wild-type mice. All septic mice were bacteremic, but no differences in bacterial load were identified between wild-type and Vil-Cre/Ikkß mice. To determine the functional significance of these results, animals were followed for survival. Septic wild-type mice had lower mortality than septic Vil-Cre/Ikkß mice (47% vs 80%, p<0.05). Antitumor necrosis factor administration decreased intestinal apoptosis, permeability, and mortality in wild-type septic mice, and a similar improvement in intestinal integrity and survival were seen when antitumor necrosis factor was given to Vil-Cre/Ikkß mice. CONCLUSIONS: Enterocyte-specific NF-kB has a beneficial role in sepsis by partially preventing sepsis-induced increases in apoptosis and permeability, which are associated with worsening mortality.


Subject(s)
Enterocytes/metabolism , I-kappa B Kinase/metabolism , Intestinal Mucosa/metabolism , Sepsis/metabolism , Sepsis/mortality , Animals , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Intestinal Mucosa/pathology , Mice , Models, Animal , Prospective Studies , Random Allocation , Tumor Necrosis Factors/pharmacology
5.
Anesthesiology ; 119(1): 166-77, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23571641

ABSTRACT

BACKGROUND: Recent clinical trials indicate that probiotic administration in critical illness has potential to reduce nosocomial infections and improve clinical outcome. However, the mechanism(s) of probiotic-mediated protection against infection and sepsis remain elusive. The authors evaluated the effects of Lactobacillus rhamnosus GG (LGG) and Bifidobacterium longum (BL) on mortality, bacterial translocation, intestinal epithelial homeostasis, and inflammatory response in experimental model of septic peritonitis. METHODS: Cecal ligation and puncture (n=14 per group) or sham laparotomy (n=8 per group) were performed on 3-week-old FVB/N weanling mice treated concomitantly with LGG, BL, or vehicle (orally gavaged). At 24 h, blood and colonic tissue were collected. In survival studies, mice were given probiotics every 24 h for 7 days (LGG, n=14; BL, n=10; or vehicle, n=13; shams, n=3 per group). RESULTS: Probiotics significantly improved mortality after sepsis (92 vs. 57% mortality for LGG and 92 vs. 50% mortality for BL; P=0.003). Bacteremia was markedly reduced in septic mice treated with either probiotic compared with vehicle treatment (4.39±0.56 vs. 1.07±1.54; P=0.0001 for LGG; vs. 2.70±1.89; P=0.016 for BL; data are expressed as mean±SD). Sepsis in untreated mice increased colonic apoptosis and reduced colonic proliferation. Probiotics significantly reduced markers of colonic apoptosis and returned colonic proliferation to sham levels. Probiotics led to significant reductions in systemic and colonic inflammatory cytokine expression versus septic animals. Our data suggest that involvement of the protein kinase B pathway (via AKT) and down-regulation of Toll-like receptor 2/Toll-like receptor 4 via MyD88 in the colon may play mechanistic roles in the observed probiotic benefits. CONCLUSIONS: Our data demonstrate that probiotic administration at initiation of sepsis can improve survival in pediatric experimental sepsis. The mechanism of this protection involves prevention of systemic bacteremia, perhaps via improved intestinal epithelial homeostasis, and attenuation of the local and systemic inflammatory responses.


Subject(s)
Homeostasis/drug effects , Intestinal Mucosa/physiology , Probiotics/therapeutic use , Sepsis/mortality , Sepsis/physiopathology , Animals , Apoptosis/drug effects , Bacteremia , Bifidobacterium , Blotting, Western , Cell Proliferation/drug effects , Colon/microbiology , Immunohistochemistry , Interleukin-6/blood , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Lacticaseibacillus rhamnosus , Mice , Oncogene Protein v-akt/metabolism , Peritonitis/drug therapy , Peritonitis/microbiology , Peritonitis/physiopathology , Proto-Oncogene Proteins c-akt/physiology , RNA/biosynthesis , RNA/genetics , Real-Time Polymerase Chain Reaction , Sepsis/microbiology , Toll-Like Receptors/physiology , Tumor Necrosis Factor-alpha/blood
6.
PLoS One ; 7(11): e49159, 2012.
Article in English | MEDLINE | ID: mdl-23145105

ABSTRACT

BACKGROUND: The small intestine plays a crucial role in the pathophysiology of sepsis and has been referred to as the "motor" of the systemic inflammatory response. One proposed mechanism is that toxic gut-derived lipid factors, transported in mesenteric lymph, induce systemic injury and distant organ failure. However, the pathways involved are yet to be defined and the role of intestinal chylomicron assembly and secretion in transporting these lipid factors is unknown. Here we studied the outcome of sepsis in mice with conditional, intestine-specific deletion of microsomal triglyceride transfer protein (Mttp-IKO), which exhibit a block in chylomicron assembly together with lipid malabsorption. METHODOLOGY/PRINCIPAL FINDINGS: Mttp-IKO mice and controls underwent intratracheal injection with either Pseudomonas aeruginosa or sterile saline. Mttp-IKO mice exhibited decreased seven-day mortality, with 0/20 (0%) dying compared to 5/17 (29%) control mice (p<0.05). This survival advantage in Mttp-IKO mice, however, was not associated with improvements in pulmonary bacterial clearance or neutrophil infiltration. Rather, Mttp-IKO mice exhibited protection against sepsis-associated decreases in villus length and intestinal proliferation and were also protected against increased intestinal apoptosis, both central features in control septic mice. Serum IL-6 levels, a major predictor of mortality in human and mouse models of sepsis, were elevated 8-fold in septic control mice but remained unaltered in septic Mttp-IKO mice. Serum high density lipoprotein (HDL) levels were reduced in septic control mice but were increased in septic Mttp-IKO mice. The decreased levels of HDL were associated with decreased hepatic expression of apolipoprotein A1 in septic control mice. CONCLUSIONS/SIGNIFICANCE: These studies suggest that strategies directed at blocking intestinal chylomicron secretion may attenuate the progression and improve the outcome of sepsis through effects mediated by metabolic and physiological adaptations in both intestinal and hepatic lipid flux.


Subject(s)
Carrier Proteins/genetics , Intestinal Diseases , Intestines/injuries , Pneumonia , Pseudomonas aeruginosa , Animals , Apoptosis , Chylomicron Remnants/metabolism , Disease Models, Animal , Female , Gene Deletion , Humans , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Diseases/physiopathology , Intestinal Mucosa/metabolism , Intestines/physiopathology , Male , Mice , Pneumonia/genetics , Pneumonia/mortality , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Sepsis/complications
7.
Shock ; 38(5): 508-14, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23042193

ABSTRACT

The endogenous bacteria have been hypothesized to play a significant role in the pathophysiology of critical illness, although their role in sepsis is poorly understood. The purpose of this study was to determine how commensal bacteria alter the host response to sepsis. Conventional and germ-free (GF) C57Bl/6 mice were subjected to Pseudomonas aeruginosa pneumonia. All GF mice died within 2 days, whereas 44% of conventional mice survived for 7 days (P = 0.001). Diluting the dose of bacteria 10-fold in GF mice led to similar survival in GF and conventional mice. When animals with similar mortality were assayed for intestinal integrity, GF mice had lower levels of intestinal epithelial apoptosis but similar levels of proliferation and intestinal permeability. Germ-free mice had significantly lower levels of tumor necrosis factor and interleukin 1ß in bronchoalveolar lavage fluid compared with conventional mice without changes in systemic cytokine production. Under conventional conditions, sepsis unmasks lymphocyte control of intestinal epithelial apoptosis, because sepsis induces a greater increase in gut apoptosis in Rag-1 mice than in wild-type mice. However, in a separate set of experiments, gut apoptosis was similar between septic GF Rag-1 mice and septic GF wild-type mice. These data demonstrate that the endogenous bacteria play a protective role in mediating mortality from pneumonia-induced sepsis, potentially mediated through altered intestinal apoptosis and the local proinflammatory response. In addition, sepsis-induced lymphocyte-dependent increases in gut epithelial apoptosis appear to be mediated by the endogenous bacteria.


Subject(s)
Apoptosis , Germ-Free Life , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa , Animals , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/pathology , Pseudomonas Infections/genetics , Pseudomonas Infections/pathology , Sepsis/genetics , Sepsis/metabolism , Sepsis/microbiology , Sepsis/pathology
8.
Shock ; 38(1): 68-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22592747

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) pneumonia-induced sepsis is a common cause of morbidity in the intensive care unit. Although pneumonia is initiated in the lungs, extrapulmonary manifestations occur commonly. In light of the key role the intestine plays in the pathophysiology of sepsis, we sought to determine whether MRSA pneumonia induces intestinal injury. FVB/N mice were subjected to MRSA or sham pneumonia and killed 24 h later. Septic animals had a marked increase in intestinal epithelial apoptosis by both hematoxylin-eosin and active caspase 3 staining. Methicillin-resistant S. aureus-induced intestinal apoptosis was associated with an increase in the expression of the proapoptotic proteins Bid and Bax and the antiapoptotic protein Bcl-xL in the mitochondrial pathway. In the receptor-mediated pathway, MRSA pneumonia induced an increase in Fas ligand but decreased protein levels of Fas, FADD, pFADD, TNF-R1, and TRADD. To assess the functional significance of these changes, MRSA pneumonia was induced in mice with genetic manipulations in proteins in either the mitochondrial or receptor-mediated pathways. Both Bid-/- mice and animals with intestine-specific overexpression of Bcl-2 had decreased intestinal apoptosis compared with wild-type animals. In contrast, Fas ligand-/- mice had no alterations in apoptosis. To determine if these findings were organism-specific, similar experiments were performed in mice subjected to Pseudomonas aeruginosa pneumonia. Pseudomonas aeruginosa induced gut apoptosis, but unlike MRSA, this was associated with increased Bcl-2 and TNF-R1 and decreased Fas. Methicillin-resistant S. aureus pneumonia thus induces organism-specific changes in intestinal apoptosis via changes in both the mitochondrial and receptor-mediated pathways, although the former may be more functionally significant.


Subject(s)
Apoptosis/physiology , Intestinal Mucosa/pathology , Methicillin-Resistant Staphylococcus aureus , Pneumonia, Staphylococcal/pathology , Animals , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Proliferation , Epithelial Cells/pathology , Fas Ligand Protein/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/physiology , Permeability , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Pneumonia, Staphylococcal/metabolism , Pseudomonas Infections/metabolism , Pseudomonas Infections/pathology , Pseudomonas aeruginosa , Sepsis/metabolism , Sepsis/microbiology , Sepsis/pathology , bcl-2-Associated X Protein/metabolism
9.
Shock ; 37(1): 85-94, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21937950

ABSTRACT

Mortality in the intensive care unit frequently results from the synergistic effect of two temporally distinct infections. This study examined the pathophysiology of a new model of intra-abdominal sepsis followed by methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. Mice underwent cecal ligation and puncture (CLP) or sham laparotomy followed 3 days later by an intratracheal injection of MRSA or saline. Both CLP/saline and sham/MRSA mice had 100% survival, whereas animals with CLP followed by MRSA pneumonia had 67% 7-day survival. Animals subjected to CLP/MRSA had increased bronchoalveolar lavage concentrations of MRSA compared with sham/MRSA animals. Animals subjected to sham/MRSA pneumonia had increased bronchoalveolar lavage levels of interleukin 6 (IL-6), tumor necrosis factor α, and granulocyte colony-stimulating factor compared with those given intratracheal saline, whereas CLP/MRSA mice had a blunted local inflammatory response with markedly decreased cytokine levels. Similarly, animals subjected to CLP/saline had increased peritoneal lavage levels of IL-6 and IL-1ß compared with those subjected to sham laparotomy, whereas this response was blunted in CLP/MRSA mice. Systemic cytokines were upregulated in both CLP/saline and sham/MRSA mice, and this was blunted by the combination of CLP/MRSA. In contrast, no synergistic effect on pneumonia severity, white blood cell count, or lymphocyte apoptosis was identified in CLP/MRSA mice compared with animals with either insult in isolation. These results indicate that a clinically relevant model of CLP followed by MRSA pneumonia causes higher mortality than could have been predicted from studying either infection in isolation, and this was associated with a blunted local (pulmonary and peritoneal) and systemic inflammatory response and decreased ability to clear infection.


Subject(s)
Cytokines/chemistry , Methicillin-Resistant Staphylococcus aureus , Pneumonia, Staphylococcal/blood , Sepsis/blood , Animals , Apoptosis/immunology , Bronchoalveolar Lavage , Cytokines/immunology , Disease Models, Animal , Leukocyte Count , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/pathology , Sepsis/immunology , Sepsis/pathology
10.
J Immunol ; 187(4): 1950-6, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21734077

ABSTRACT

Lymphocyte apoptosis is thought to have a major role in the pathophysiology of sepsis. However, there is a disconnect between animal models of sepsis and patients with the disease, because the former use subjects that were healthy prior to the onset of infection while most patients have underlying comorbidities. The purpose of this study was to determine whether lymphocyte apoptosis prevention is effective in preventing mortality in septic mice with preexisting cancer. Mice with lymphocyte Bcl-2 overexpression (Bcl-2-Ig) and wild type (WT) mice were injected with a transplantable pancreatic adenocarcinoma cell line. Three weeks later, after development of palpable tumors, all animals received an intratracheal injection of Pseudomonas aeruginosa. Despite having decreased sepsis-induced T and B lymphocyte apoptosis, Bcl-2-Ig mice had markedly increased mortality compared with WT mice following P. aeruginosa pneumonia (85 versus 44% 7-d mortality; p = 0.004). The worsened survival in Bcl-2-Ig mice was associated with increases in Th1 cytokines TNF-α and IFN-γ in bronchoalveolar lavage fluid and decreased production of the Th2 cytokine IL-10 in stimulated splenocytes. There were no differences in tumor size or pulmonary pathology between Bcl-2-Ig and WT mice. To verify that the mortality difference was not specific to Bcl-2 overexpression, similar experiments were performed in Bim(-/-) mice. Septic Bim(-/-) mice with cancer also had increased mortality compared with septic WT mice with cancer. These data demonstrate that, despite overwhelming evidence that prevention of lymphocyte apoptosis is beneficial in septic hosts without comorbidities, the same strategy worsens survival in mice with cancer that are given pneumonia.


Subject(s)
Adenocarcinoma/immunology , Apoptosis/immunology , Lymphocytes/immunology , Pancreatic Neoplasms/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Sepsis/immunology , Adenocarcinoma/genetics , Adenocarcinoma/microbiology , Animals , Apoptosis/genetics , Cytokines/biosynthesis , Cytokines/immunology , Humans , Mice , Mice, Knockout , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/microbiology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Proto-Oncogene Proteins c-bcl-2/genetics , Pseudomonas Infections/microbiology , Sepsis/genetics , Sepsis/microbiology
11.
Shock ; 36(4): 381-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21701422

ABSTRACT

Mortality from pneumonia is mediated, in part, through extrapulmonary causes. Epidermal growth factor (EGF) has broad cytoprotective effects, including potent restorative properties in the injured intestine. The purpose of this study was to determine the efficacy of EGF treatment following Pseudomonas aeruginosa pneumonia. FVB/N mice underwent intratracheal injection of either P. aeruginosa or saline and were then randomized to receive either systemic EGF or vehicle beginning immediately or 24 h after the onset of pneumonia. Systemic EGF decreased 7-day mortality from 65% to 10% when initiated immediately after the onset of pneumonia and to 27% when initiated 24 h after the onset of pneumonia. Even though injury in pneumonia is initiated in the lungs, the survival advantage conferred by EGF was not associated with improvements in pulmonary pathology. In contrast, EGF prevented intestinal injury by reversing pneumonia-induced increases in intestinal epithelial apoptosis and decreases in intestinal proliferation and villus length. Systemic cytokines and kidney and liver function were unaffected by EGF therapy, although EGF decreased pneumonia-induced splenocyte apoptosis. To determine whether the intestine was sufficient to account for extrapulmonary effects induced by EGF, a separate set of experiments was done using transgenic mice with enterocyte-specific overexpression of EGF (IFABP-EGF [intestinal fatty acid-binding protein linked to mouse EGF] mice), which were compared with wild-type mice subjected to pneumonia. IFABP-EGF mice had improved survival compared with wild-type mice following pneumonia (50% vs. 28%, respectively, P < 0.05) and were protected from pneumonia-induced intestinal injury. Thus, EGF may be a potential adjunctive therapy for pneumonia, mediated in part by its effects on the intestine.


Subject(s)
Epidermal Growth Factor/metabolism , Intestinal Diseases/prevention & control , Pneumonia/drug therapy , Pseudomonas aeruginosa/pathogenicity , Animals , Apoptosis/drug effects , Disease Models, Animal , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestines/drug effects , Intestines/pathology , Mice , Mice, Transgenic , Peroxidase/metabolism , Pneumonia/complications , Pneumonia/microbiology
12.
J Immunol ; 186(6): 3718-25, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21296977

ABSTRACT

Sepsis is primarily a disease of the aged, with increased incidence and mortality occurring in aged hosts. Heat shock protein (HSP) 70 plays an important role in both healthy aging and the stress response to injury. The purpose of this study was to determine the role of HSP70 in mediating mortality and the host inflammatory response in aged septic hosts. Sepsis was induced in both young (6- to 12-wk-old) and aged (16- to 17-mo-old) HSP70(-/-) and wild-type (WT) mice to determine whether HSP70 modulated outcome in an age-dependent fashion. Young HSP70(-/-) and WT mice subjected to cecal ligation and puncture, Pseudomonas aeruginosa pneumonia, or Streptococcus pneumoniae pneumonia had no differences in mortality, suggesting HSP70 does not mediate survival in young septic hosts. In contrast, mortality was higher in aged HSP70(-/-) mice than aged WT mice subjected to cecal ligation and puncture (p = 0.01), suggesting HSP70 mediates mortality in sepsis in an age-dependent fashion. Compared with WT mice, aged septic HSP70(-/-) mice had increased gut epithelial apoptosis and pulmonary inflammation. In addition, HSP70(-/-) mice had increased systemic levels of TNF-α, IL-6, IL-10, and IL-1ß compared with WT mice. These data demonstrate that HSP70 is a key determinant of mortality in aged, but not young hosts in sepsis. HSP70 may play a protective role in an age-dependent response to sepsis by preventing excessive gut apoptosis and both pulmonary and systemic inflammation.


Subject(s)
Aging/immunology , Disease Models, Animal , HSP70 Heat-Shock Proteins/physiology , Sepsis/immunology , Sepsis/mortality , Aging/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Cecum , Female , HSP70 Heat-Shock Proteins/deficiency , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Ligation , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/pathology , Pseudomonas Infections/immunology , Pseudomonas Infections/mortality , Pseudomonas Infections/pathology , Punctures , Sepsis/pathology , Streptococcal Infections/immunology , Streptococcal Infections/mortality , Streptococcal Infections/pathology
14.
Crit Care Clin ; 26(3): 549-65, x, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20643306

ABSTRACT

The intestine plays a central role in the pathophysiology of critical illness and is frequently called the "motor" of the systemic inflammatory response. Perturbations to the intestinal barrier can lead to distant organ damage and multiple organ failure. Therefore, identifying ways to preserve intestinal integrity may be of paramount importance. Growth factors and other peptides have emerged as potential tools for modulation of intestinal inflammation and repair due to their roles in cellular proliferation, differentiation, migration, and survival. This review examines the involvement of growth factors and other peptides in intestinal epithelial repair during critical illness and their potential use as therapeutic targets.


Subject(s)
Critical Illness/therapy , Gastrointestinal Tract/drug effects , Intercellular Signaling Peptides and Proteins/therapeutic use , Systemic Inflammatory Response Syndrome/prevention & control , Critical Care/methods , Gastrointestinal Tract/metabolism , Humans , Intercellular Signaling Peptides and Proteins/physiology , Systemic Inflammatory Response Syndrome/metabolism
15.
Crit Care Med ; 38(3): 886-93, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20009755

ABSTRACT

OBJECTIVE: Whereas most septic patients have an underlying comorbidity, most animal models of sepsis use mice that were healthy before the onset of infection. Malignancy is the most common comorbidity associated with sepsis. The purpose of this study was to determine whether mice with cancer have a different response to sepsis than healthy animals. DESIGN: Prospective, randomized controlled study. SETTING: Animal laboratory in a university medical center. SUBJECTS: C57Bl/6 mice. INTERVENTIONS: Animals received a subcutaneous injection of either 250,000 cells of the transplantable pancreatic adenocarcinoma cell line Pan02 (cancer) or phosphate-buffered saline (healthy). Three weeks later, mice given Pan02 cells had reproducible, nonmetastatic tumors. Both groups of mice then underwent intratracheal injection of either Pseudomonas aeruginosa (septic) or 0.9% NaCl (sham). Animals were killed 24 hrs postoperatively or followed-up 7 days for survival. MEASUREMENTS AND MAIN RESULTS: Mice with cancer and healthy mice appeared similar when subjected to sham operation, although cancer animals had lower levels of T- and B-lymphocyte apoptosis. Septic mice with cancer had increased mortality compared to previously healthy septic mice subjected to the identical injury (52% vs. 28%; p = .04). This was associated with increased bacteremia but no difference in local pulmonary infection. Septic mice with cancer also had increased intestinal epithelial apoptosis. Although sepsis induced an increase in T- and B-lymphocyte apoptosis in all animals, septic mice with cancer had decreased T- and B-lymphocyte apoptosis compared to previously healthy septic mice. Serum and pulmonary cytokines, lung histology, complete blood counts, and intestinal proliferation were similar between septic mice with cancer and previously healthy septic mice. CONCLUSIONS: When subjected to the same septic insult, mice with cancer have increased mortality compared to previously healthy animals. Decreased systemic bacterial clearance and alterations in intestinal epithelial and lymphocyte apoptosis may help explain this differential response.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/pathology , Apoptosis/physiology , Bacteremia/mortality , Bacteremia/pathology , Disease Models, Animal , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa , Sepsis/mortality , Sepsis/pathology , Animals , B-Lymphocytes/pathology , Cell Line, Tumor , Comorbidity , Intestinal Mucosa/pathology , Lung/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Pseudomonas Infections/mortality , T-Lymphocytes/pathology
18.
Pediatr Res ; 53(3): 426-33, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12595590

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating intestinal disease of premature infants. Maternal milk has been suggested to be partially protective against NEC; however, the mechanisms of this protection are not defined. The aim of this study was to examine the effect(s) of artificial feeding of rat milk (RM)-versus cow milk-based rat milk substitute (RMS) on the development of NEC in a neonatal rat model and elucidate the role of inflammatory cytokines in NEC pathogenesis. Newborn rats were artificially fed with either collected RM or RMS. Experimental NEC was induced by exposure to asphyxia and cold stress and evaluated by histologic scoring of damage in ileum. Intestinal cytokine mRNA expression was determined by real-time PCR. Cytokine histologic localization was performed by confocal microscopy. Similar to human NEC, artificial feeding of RM reduces the incidence and severity of NEC injury in neonatal rats. Freezing and thawing of collected RM did not eliminate the protective effect of maternal milk. Ileal IL-10 expression was significantly increased in the RM group compared with RMS. Increased IL-10 peptide production was detected in the RM group with signal localized predominantly in the cytoplasm of villus epithelial cells. These results suggest that the protective effect of maternal milk is associated with increased production of anti-inflammatory IL-10 in the site of injury. Better understanding of the mechanisms underlying these protective effects could be beneficial either in the prevention of NEC or in the development of future therapeutic strategies to cure NEC.


Subject(s)
Enterocolitis, Necrotizing/diet therapy , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Milk , Animals , Animals, Newborn , Asphyxia/immunology , Asphyxia/metabolism , Cold Temperature , Disease Models, Animal , Enteral Nutrition , Enterocolitis, Necrotizing/epidemiology , Enterocolitis, Necrotizing/immunology , Female , Ileum/immunology , Ileum/metabolism , Ileum/ultrastructure , Incidence , Intestinal Mucosa/immunology , Intestinal Mucosa/ultrastructure , Microscopy, Electron, Scanning , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Stress, Physiological/immunology , Stress, Physiological/metabolism , Weight Gain
19.
Am J Physiol Gastrointest Liver Physiol ; 284(4): G695-702, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12529262

ABSTRACT

Necrotizing enterocolitis (NEC) is a common and devastating gastrointestinal disease of premature infants. Along with pathological effects in the ileum, severe NEC is often accompanied by multisystem organ failure, including liver failure. The aim of this study was to determine the changes in hepatic cytokines and inflammatory mediators in experimental NEC. The well-established neonatal rat model of NEC was used in this study, and changes in liver morphology, numbers of Kupffer cells (KC), gene expression, and histological localization of IL-18, TNF-alpha, and inducible nitric oxide synthase were evaluated. Intestinal luminal TNF-alpha levels were also measured. Production of hepatic IL-18 and TNF-alpha and numbers of KC were increased in rats with NEC and correlated with the progression of intestinal damage during NEC development. Furthermore, increased levels of TNF-alpha in the intestinal lumen of rats with NEC was significantly decreased when KC were inhibited with gadolinium chloride. These results suggest an important role of the liver and the gut-liver axis in NEC pathogenesis.


Subject(s)
Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/physiopathology , Inflammation Mediators/metabolism , Liver/immunology , Tyrosine/analogs & derivatives , Animals , Animals, Newborn , Asphyxia/immunology , Cold Temperature , Enteral Nutrition , Enterocolitis, Necrotizing/pathology , Gene Expression/immunology , Interleukin-18/genetics , Interleukin-18/metabolism , Intestines/immunology , Kupffer Cells/pathology , Liver/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Stress, Physiological/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/metabolism
20.
J Pediatr Gastroenterol Nutr ; 36(1): 126-33, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12500008

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is the most common gastrointestinal disease of premature infants. We have shown in previous studies that proinflammatory interleukin-18 and interleukin-12 are up-regulated in the ileum of rats with experimental NEC and that epidermal growth factor (EGF) reduces the development of disease. Here we investigated whether the protective effects of EGF are a result of changes in ileal interleukin-18, interleukin-12 and/or antiinflammatory interleukin-10. METHODS: Newborn rats were artificially fed with either growth-factor-free rat milk substitute (RMS) or RMS supplemented with 500 ng/mL EGF (RMS + EGF) and NEC was induced via exposure to asphyxia and cold stress. Cytokine expression and localization were assessed using reverse-transcription real-time polymerase chain reaction and immunohistology/confocal microscopy. RESULTS: Enteral administration of EGF (RMS + EGF) decreased overproduction of interleukin-18 and increased interleukin-10 production in the ileum. Furthermore, increased interleukin-10 production was associated with up-regulation of the transcription factor Sp1 in RMS + EGF rats. CONCLUSIONS: These data suggest that EGF may reduce NEC via increased interleukin-10 and decreased interleukin-18 and that EGF-mediated up-regulation of Sp1 may account for the increased interleukin-10.


Subject(s)
Cytokines/drug effects , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Epidermal Growth Factor/pharmacology , Ileum/immunology , Ileum/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Immunohistochemistry , Interleukins/metabolism , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL