Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 13: 1298360, 2023.
Article in English | MEDLINE | ID: mdl-38089821

ABSTRACT

Background: Neisseria meningitidis (Nm) is the cause of epidemic meningitis and fulminant meningococcal septicemia. The clinical presentations and outcome of meningococcal septic shock is closely related to the circulating levels of lipopolysaccharides (LPS) and of Neisseria meningitidis DNA (Nm DNA). We have previously explored the distribution of Nm DNA in tissues from large organs of patients dying of meningococcal septic shock and in a porcine meningococcal septic shock model. Objective: 1) To explore the feasibility of measuring LPS levels in tissues from the large organs in patients with meningococcal septic shock and in a porcine meningococcal septic shock model. 2) To evaluate the extent of contamination of non-specific LPS during the preparation of tissue samples. Patients and methods: Plasma, serum, and fresh frozen (FF) tissue samples from the large organs of three patients with lethal meningococcal septic shock and two patients with lethal pneumococcal disease. Samples from a porcine meningococcal septic shock model were included. Frozen tissue samples were thawed, homogenized, and prepared for quantification of LPS by Pyrochrome® Limulus Amoebocyte Lysate (LAL) assay. Results: N. meningitidis DNA and LPS was detected in FF tissue samples from large organs in all patients with meningococcal septic shock. The lungs are the organs with the highest LPS and Nm DNA concentration followed by the heart in two of the three meningococcal shock patients. Nm DNA was not detected in any plasma or tissue sample from patients with lethal pneumococcal infection. LPS was detected at a low level in all FF tissues from the two patients with lethal pneumococcal disease. The experimental porcine meningococcal septic shock model indicates that also in porcinis the highest LPS and Nm DNA concentration are detected in lungs tissue samples. The quantification analysis showed that the highest concentration of both Nm DNA and LPS are in the organs and not in the circulation of patients with lethal meningococcal septic shock. This was also shown in the experimental porcine meningococcal septic shock model. Conclusion: Our results suggest that LPS can be quantified in mammalian tissues by using the LAL assay.


Subject(s)
Meningitis, Meningococcal , Meningococcal Infections , Neisseria meningitidis , Pneumococcal Infections , Sepsis , Shock, Septic , Animals , Humans , DNA , Lipopolysaccharides , Mammals , Swine
2.
Front Cell Infect Microbiol ; 12: 908204, 2022.
Article in English | MEDLINE | ID: mdl-36034711

ABSTRACT

Background: Fulminant meningococcal sepsis with shock and multiple organ failure is associated with a massive systemic inflammatory response involving solid organs. We have previously established a porcine model of the disease to study pathophysiologic and possible therapeutic strategies. Objective: This study examined whether the organ specific gene expression profile in such a large animal model reflects the profile seen in patients with fulminant meningococcal sepsis. Patients and methods: Data from gene expression profiles induced in organs from patients (n=5) and the porcine model (n=8) were imported into the Ingenuity pathway analysis (IPA) software for comparison analysis. The number of meningococci in the organs were quantified by real time-PCR. Results: The all-over transcriptional activation between different organs revealed a striking concordance between the patients and the pigs regarding the pattern of transcriptional activation and activated pathways. Comparison analysis demonstrated similar pattern of upregulation of genes being associated with a large range of inflammatory biofunctions in the patients and the porcine model. Genes associated with biofunctions such as organismal death, morbidity and mortality were similarly downregulated in the patients and the porcine model. Comparison analysis of main predicted canonical pathways also demonstrated a high degree of similarity regarding up- and downregulation in both groups. Core analysis revealed different top-upstream regulators in the different organs in the patients. In the patients pro-inflammatory regulators were most activated in the lungs. In the other organs up-stream factors that regulate signaling pathways involved in development, growth, repair and homeostasis and triglyceride synthesis were most activated. In the porcine model, the top-upstream regulators were pro-inflammatory in all organs. The difference may reflect the shorter duration of the porcine experiment than the duration of the patient's infection before death. Conclusion: The inflammatory responses measured on the transcriptomic level in organs in patients with fulminant meningococcal sepsis is reproduced in the porcine model of the disease, although some differences may exist regarding the top-upregulated factors in individual organs. Thus, this large animal model reproduces important immunological features of meningococcal sepsis and can be a valuable tool in further investigations of inflammatory aspects and possible treatment options.


Subject(s)
Neisseria meningitidis , Sepsis , Shock, Septic , Animals , Disease Models, Animal , Swine , Transcriptome
3.
Article in English | MEDLINE | ID: mdl-32154187

ABSTRACT

Background: Patients developing meningococcal septic shock reveal levels of Neisseria meningitidis (106-108/mL) and endotoxin (101-103 EU/mL) in the circulation and organs, leading to acute cardiovascular, pulmonary and renal failure, coagulopathy and a high case fatality rate within 24 h. Objective: To investigate transcriptional profiles in heart, lungs, kidneys, liver, and spleen and immunostain key inflammatory cells and proteins in post mortem formalin-fixed, paraffin-embedded (FFPE) tissue samples from meningococcal septic shock patients. Patients and Methods: Total RNA was isolated from FFPE and fresh frozen (FF) tissue samples from five patients and two controls (acute non-infectious death). Differential expression of genes was detected using Affymetrix microarray analysis. Lung and heart tissue samples were immunostained for T-and B cells, macrophages, neutrophils and the inflammatory markers PAI-1 and MCP-1. Inflammatory mediators were quantified in lysates from FF tissues. Results: The transcriptional profiles showed a complex pattern of protein-coding and non-coding RNAs with significant regulation of pathways associated with organismal death, cell death and survival, leukocyte migration, cellular movement, proliferation of cells, cell-to-cell signaling, immune cell trafficking, and inflammatory responses in an organ-specific clustering manner. The canonical pathways including acute phase response-, EIF2-, TREM1-, IL-6-, HMBG1-, PPAR signaling, and LXR/RXR activation were associated with acute heart, pulmonary, and renal failure. Fewer genes were regulated in the liver and particularly in the spleen. The main upstream regulators were TNF, IL-1ß, IL-6, RICTOR, miR-6739-3p, and CD3. Increased numbers of inflammatory cells (CD68+, MPO+, CD3+, and CD20+) were found in lungs and heart. PAI-1 inhibiting fibrinolysis and MCP-1 attracting leukocyte were found significantly present in the septic tissue samples compared to the controls. Conclusions: FFPE tissue samples can be suitable for gene expression studies as well as immunostaining of specific cells or molecules. The most pronounced gene expression patterns were found in the organs with highest levels of Neisseria meningitidis DNA. Thousands of protein-coding and non-coding RNA transcripts were altered in lungs, heart and kidneys. We identified specific biomarker panels both protein-coding and non-coding RNA transcripts, which differed from organ to organ. Involvement of many genes and pathways add up and the combined effect induce organ failure.


Subject(s)
Neisseria meningitidis , Shock, Septic , Gene Expression Profiling , Humans , Multiple Organ Failure , Transcriptome
4.
BMC Clin Pathol ; 17: 10, 2017.
Article in English | MEDLINE | ID: mdl-28824331

ABSTRACT

BACKGROUND: The pathophysiology and outcome of meningococcal septic shock is closely associated with the plasma level of N. meningitidis lipopolysaccharides (LPS, endotoxin) and the circulating level of meningococcal DNA. The aim of the present study was to quantify the number of N. meningitidis in different formalin-fixed, paraffin-embedded (FFPE) tissue samples and fresh frozen (FF) tissue samples from patients with systemic meningococcal disease (SMD), to explore the distribution of N. meningitidis in the body. METHODS: DNA in FFPE and FF tissue samples from heart, lungs, liver, kidneys, spleen and brain from patients with meningococcal shock and controls (lethal pneumococcal infection) stored at variable times, were isolated. The bacterial load of N. meningitidis DNA was analyzed using quantitative real-time PCR (qPCR) and primers for the capsule transport A (ctrA) gene (1 copy per N. meningitidis DNA). The human beta-hemoglobin (HBB) gene was quantified to evaluate effect of the storage times (2-28 years) and storage method in archived tissue. RESULTS: N. meningitidis DNA was detected in FFPE and FF tissue samples from heart, lung, liver, kidney, and spleen in all patients with severe shock. In FFPE brain, N. meningitidis DNA was only detected in the patient with the highest concentration of LPS in the blood at admission to hospital. The highest levels of N. meningitidis DNA were found in heart tissue (median value 3.6 × 107 copies N. meningitidis DNA/µg human DNA) and lung tissue (median value 3.1 × 107 copies N. meningitidis DNA/µg human DNA) in all five patients. N. meningitidis DNA was not detectable in any of the tissue samples from two patients with clinical meningitis and the controls (pneumococcal infection). The quantity of HBB declined over time in FFPE tissue stored at room temperature, suggesting degradation of DNA. CONCLUSIONS: High levels of N. meningitidis DNA were detected in the different tissue samples from meningococcal shock patients, particularly in the heart and lungs suggesting seeding and major proliferation of meningococci in these organs during the development of shock, probably contributing to the multiple organ failure. The age of archived tissue samples appear to have an impact on the amount of quantifiable N. meningitidis DNA.

5.
Proc Natl Acad Sci U S A ; 114(4): E534-E539, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069958

ABSTRACT

Two functions have been assigned to properdin; stabilization of the alternative convertase, C3bBb, is well accepted, whereas the role of properdin as pattern recognition molecule is controversial. The presence of nonphysiological aggregates in purified properdin preparations and experimental models that do not allow discrimination between the initial binding of properdin and binding secondary to C3b deposition is a critical factor contributing to this controversy. In previous work, by inhibiting C3, we showed that properdin binding to zymosan and Escherichia coli is not a primary event, but rather is solely dependent on initial C3 deposition. In the present study, we found that properdin in human serum bound dose-dependently to solid-phase myeloperoxidase. This binding was dependent on C3 activation, as demonstrated by the lack of binding in human serum with the C3-inhibitor compstatin Cp40, in C3-depleted human serum, or when purified properdin is applied in buffer. Similarly, binding of properdin to the surface of human umbilical vein endothelial cells or Neisseria meningitidis after incubation with human serum was completely C3-dependent, as detected by flow cytometry. Properdin, which lacks the structural homology shared by other complement pattern recognition molecules and has its major function in stabilizing the C3bBb convertase, was found to bind both exogenous and endogenous molecular patterns in a completely C3-dependent manner. We therefore challenge the view of properdin as a pattern recognition molecule, and argue that the experimental conditions used to test this hypothesis should be carefully considered, with emphasis on controlling initial C3 activation under physiological conditions.


Subject(s)
Complement C3b/metabolism , Properdin/metabolism , Cells, Cultured , Complement Activation , Granulocytes/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neisseria meningitidis , Peptides, Cyclic/pharmacology , Peroxidase/metabolism , Serum
6.
Infect Immun ; 81(9): 3173-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23774598

ABSTRACT

Sepsis is an infection-induced systemic inflammatory response syndrome. Upstream recognition molecules, like CD14, play key roles in the pathogenesis. The aim of the present study was to investigate the effect of systemic CD14 inhibition on local inflammatory responses in organs from septic pigs. Pigs (n = 34) receiving Escherichia coli-bacteria or E. coli-lipopolysaccharide (LPS) were treated with an anti-CD14 monoclonal antibody or an isotype-matched control. Lungs, liver, spleen, and kidneys were examined for bacteria and inflammatory biomarkers. E. coli and LPS were found in large amounts in the lungs compared to the liver, spleen, and kidneys. Notably, the bacterial load did not predict the respective organ inflammatory response. There was a marked variation in biomarker induction in the organs and in the effect of anti-CD14. Generally, the spleen produced the most cytokines per weight unit, whereas the liver contributed the most to the total load. All cytokines were significantly inhibited in the spleen. Interleukin-6 (IL-6) was significantly inhibited in all organs, IL-1ß and IP-10 were significantly inhibited in liver, spleen, and kidneys, and tumor necrosis factor, IL-8, and PAI-1 were inhibited only in the spleen. ICAM-1 and VCAM-1 was significantly inhibited in the kidneys. Systemic CD14-inhibition efficiently, though organ dependent, attenuated local inflammatory responses. Detailed knowledge on how the different organs respond to systemic inflammation in vivo, beyond the information gained by blood examination, is important for our understanding of the nature of systemic inflammation and is required for future mediator-directed therapy in sepsis. Inhibition of CD14 seems to be a good candidate for such treatment.


Subject(s)
Escherichia coli/immunology , Inflammation/immunology , Lipopolysaccharide Receptors/immunology , Sepsis/immunology , Swine/immunology , Animal Structures/immunology , Animal Structures/metabolism , Animals , Antibodies, Monoclonal/immunology , Biomarkers/metabolism , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Escherichia coli/metabolism , Inflammation/metabolism , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interleukins/immunology , Interleukins/metabolism , Lipopolysaccharide Receptors/metabolism , Plasminogen Activator Inhibitor 1/immunology , Plasminogen Activator Inhibitor 1/metabolism , Sepsis/metabolism , Swine/metabolism , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
7.
Immunobiology ; 218(5): 683-9, 2013 May.
Article in English | MEDLINE | ID: mdl-22947599

ABSTRACT

Evidence suggests that adjunctive treatment with intravenous immunoglobulin preparations enriched with IgA and IgM reduce mortality in sepsis. The mode of action of polyvalent immunoglobulin is complex, including neutralization of toxins and modulation of complement activation and cytokine formation toward an anti-inflammatory profile. In this study we explored the effect of Pentaglobin, containing IgG, IgA and IgM, on the initial inflammatory reaction as well as on hemodynamics, using a well characterized and standardized porcine model of sepsis. Anesthetized and mechanically ventilated pigs, mean weight 14.9 kg, were allocated into two groups of 8 animals, receiving either Pentaglobin or saline, before sepsis was induced by intravenous Escherichia coli infusion. Five negative controls received saline only. All animals were observed for 4 h under extensive invasive monitoring. Pentaglobin significantly (p < 0.05) attenuated IL-1ß formation by 38% at the end of the experiment, and markedly increased (p < 0.05) the formation of IL-10 at 60 min. TNF-α, IL-6, IL-8 and expression of the cell surface marker wCD11R3 were lower in the Pentaglobin group, but the differences were not significant. The serum concentration of LPS was three times higher in the Pentaglobin group (p < 0.005), indicating binding of LPS to Pentaglobin. Complementary in vitro experiments showed a higher binding affinity for IgM and IgA to LPS than for IgG. LPS-induced formation of IL-6 was significantly (p < 0.05) attenuated by Pentaglobin in an in vitro whole blood model. In conclusion, Pentaglobin decreased the key inflammasome IL-1ß molecule in an E. coli-model of pigs sepsis.


Subject(s)
Escherichia coli Infections/drug therapy , Immunoglobulin A/pharmacology , Immunoglobulin M/pharmacology , Immunoglobulins, Intravenous/pharmacology , Interleukin-1beta/antagonists & inhibitors , Sepsis/drug therapy , Animals , Antigen-Antibody Complex/blood , Biomarkers/blood , Escherichia coli/drug effects , Escherichia coli/immunology , Escherichia coli Infections/blood , Escherichia coli Infections/immunology , Hemodynamics/drug effects , Immunoglobulin A/blood , Immunoglobulin M/blood , Immunoglobulins, Intravenous/blood , Inflammation/prevention & control , Interleukin-10/blood , Interleukin-10/immunology , Interleukin-1beta/blood , Interleukin-1beta/immunology , Interleukin-6/blood , Interleukin-6/immunology , Interleukin-8/blood , Interleukin-8/immunology , Lipopolysaccharides/blood , Protein Binding , Sepsis/blood , Sepsis/immunology , Swine , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
8.
Shock ; 36(5): 517-23, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21921836

ABSTRACT

Bradykinin (BK) is regarded as an important mediator of edema, shock, and inflammation during sepsis. In this study, we evaluated the contribution of BK in porcine sepsis by blocking BK and by measuring the stable BK metabolite, BK1-5, using anesthetized pigs. The effect of BK alone, the efficacy of icatibant to block this effect, and the recovery of BK measured as plasma BK1-5 were first investigated. Purified BK injected intravenously induced an abrupt fall in blood pressure, which was completely prevented by pretreatment with icatibant. BK1-5 was detected in plasma corresponding to the doses given. The effect of icatibant was then investigated in an established model of porcine gram-negative sepsis. Neisseria meningitidis was infused intravenously without any pretreatment (n = 8) or pretreated with icatibant (n = 8). Negative controls received saline only. Icatibant-treated pigs developed the same degree of severe sepsis as did the controls. Both groups had massive capillary leakage, leukopenia, and excessive cytokine release. The plasma level of BK1-5 was low or nondetectable in all pigs. The latter observation was confirmed in supplementary studies with pigs undergoing Escherichia coli or polymicrobial sepsis induced by cecal ligation and puncture. In conclusion, icatibant completely blocked the hemodynamic effects of BK but had no beneficial effects on N. meningitidis-induced edema, shock, and inflammation. This and the fact that plasma BK1-5 in all the septic pigs was virtually nondetectable question the role of BK as an important mediator of porcine sepsis. Thus, the data challenge the current view of the role of BK also in human sepsis.


Subject(s)
Bradykinin Receptor Antagonists , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Bradykinin/pharmacology , Sepsis/metabolism , Animals , Bradykinin/therapeutic use , Edema/drug therapy , Edema/microbiology , Inflammation/drug therapy , Inflammation/microbiology , Neisseria meningitidis/pathogenicity , Sepsis/drug therapy , Shock/drug therapy , Shock/microbiology , Swine
9.
Mol Immunol ; 48(15-16): 2159-69, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21839519

ABSTRACT

The initial interaction of Gram-negative bacteria with erythrocytes and its implications on leukocyte phagocytosis and oxidative burst in human whole blood were examined. Alexa-labeled Escherichia coli, wild-type H44/76 N. meningitidis and the H44/76lpxA lipopolysaccharide (LPS)-deficient mutant were incubated with whole blood using lepirudin as anticoagulant which has no adverse effects on complement. Bacteria free in plasma, bound to erythrocytes or phagocytized by granulocytes and monocytes were quantified using flow cytometry. The effects of the C3 inhibitor compstatin, a C5a receptor antagonist (C5aRa) and a complement receptor 1 (CR1)-blocking antibody (3D9) were examined. Most bacteria (80%) immediately bound to erythrocytes. The binding gradually declined over time, with a parallel increase in phagocytosis. Complement inhibition with compstatin reduced erythrocyte binding and bacterial C3 opsonization. In contrast, the C5aRa efficiently reduced phagocytosis, but did not affect the binding of bacteria to erythrocytes. The anti-CR1 blocking mAb dose-dependently reduced bacterial binding to erythrocytes to nil, with subsequent increased phagocytosis and oxidative burst. LPS had no effect on these processes since similar results were obtained using an LPS-deficient N. meningitidis mutant. In vivo experiments in a pig model of sepsis showed limited binding of bacteria to erythrocytes, consistent with the facts that erythrocyte CR1 receptors are absent in non-primates and that the bacteria were mainly found in the lungs. In conclusion, complement-dependent binding of Gram-negative bacteria to erythrocyte CR1 decreases phagocytosis and oxidative burst by leukocytes in human whole blood.


Subject(s)
Erythrocytes/immunology , Gram-Negative Bacterial Infections/immunology , Leukocytes/immunology , Phagocytosis/immunology , Receptors, Complement 3b/metabolism , Animals , Cell Separation , Complement System Proteins , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Erythrocytes/metabolism , Escherichia coli/immunology , Escherichia coli/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gram-Negative Bacterial Infections/metabolism , Humans , Leukocytes/metabolism , Neisseria meningitidis/immunology , Neisseria meningitidis/metabolism , Respiratory Burst , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/immunology , Sepsis/metabolism , Swine
10.
J Diabetes Sci Technol ; 5(6): 1490-8, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22226271

ABSTRACT

BACKGROUND: Continuous monitoring of glucose by implantable microfabricated devices offers key advantages over current transcutaneous glucose sensors that limit usability due to their obtrusive nature and risk of infection. A successful sensory implant should be biocompatible and retain long-lasting function. Polymorphonuclear leukocytes (PMN) play a key role in the inflammatory system by releasing enzymes, cytokines, and reactive oxygen species, typically as a response to complement activation. The aim of this study was to perform an in vitro analysis of PMN activation as a marker for biocompatibility of materials and to evaluate the role of complement in the activation of PMN. METHODS: Fifteen candidate materials of an implantable glucose sensor were incubated in lepirudin-anticoagulated whole blood. The cluster of differentiation molecule 11b (CD11b) expression on PMN was analyzed with flow cytometry and the myeloperoxidase (MPO) concentration in plasma was analyzed with enzyme-linked immunosorbent assay. Complement activation was prevented by the C3 inhibitor compstatin or the C5 inhibitor eculizumab. RESULTS: Three of the biomaterials (cellulose ester, polyamide reverse osmosis membrane, and polyamide thin film membrane), all belonging to the membrane group, induced a substantial and significant increase in CD11b expression and MPO release. The changes were virtually identical for these two markers. Inhibition of complement with compstatin or eculizumab reduced the CD11b expression and MPO release dose dependently and in most cases back to baseline. The other 12 materials did not induce significant PMN activation. CONCLUSION: Three of the 15 candidate materials triggered PMN activation in a complement-dependent manner and should therefore be avoided for implementation in implantable microsensors.


Subject(s)
Biocompatible Materials/adverse effects , Materials Testing , Neutrophils/immunology , Prostheses and Implants/adverse effects , Blood Glucose/analysis , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans
11.
Infect Immun ; 78(2): 802-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19933829

ABSTRACT

Certain complement defects are associated with an increased propensity to contract Neisseria meningitidis infections. We performed detailed analyses of complement-mediated defense mechanisms against N. meningitidis 44/76 with whole blood and serum from two adult patients who were completely C2 or C5 deficient. The C5-deficient patient and the matched control were also deficient in mannose-binding lectin (MBL). The proliferation of meningococci incubated in freshly drawn whole blood was estimated by CFU and quantitative DNA real-time PCR. The serum bactericidal activity and opsonophagocytic activity by granulocytes were investigated, including heat-inactivated postvaccination sera, to examine the influence of antimeningococcal antibodies. The meningococci proliferated equally in C2- and C5-deficient blood, with a 2 log(10) increase of CFU and 4- to 5-log(10) increase in DNA copies. Proliferation was modestly decreased in reconstituted C2-deficient and control blood. After reconstitution of C5-deficient blood, all meningococci were killed, which is consistent with high antibody titers being present. The opsonophagocytic activity was strictly C2 dependent, appeared with normal serum, and increased with postvaccination serum. Serum bactericidal activity was strictly dependent on C2, C5, and high antibody titers. MBL did not influence any of the parameters observed. Complement-mediated defense against meningococci was thus dependent on the classical pathway. Some opsonophagocytic activity occurred despite low levels of antimeningococcal antibodies but was more efficient with immune sera. Serum bactericidal activity was dependent on C2, C5, and immune sera. MBL did not influence any of the parameters observed.


Subject(s)
Antibodies/immunology , Complement C2/immunology , Complement C5/immunology , Neisseria meningitidis/immunology , Adolescent , Adult , Antibodies/blood , Complement C2/deficiency , Complement C2/genetics , Complement C5/deficiency , Complement C5/genetics , Female , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction
12.
FASEB J ; 24(3): 712-22, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19841036

ABSTRACT

Sepsis is a severe infection-induced systemic inflammatory syndrome. Inhibition of downstream inflammatory mediators of sepsis, e.g., TNF-alpha, has failed in clinical trials. The aim of this study was to investigate the effects of inhibiting CD14, a key upstream innate immunity molecule, on the early inflammatory and hemostatic responses in a pig model of gram-negative sepsis. The study comprised two arms, whole live Escherichia coli bacteria and E. coli lipopolysaccharide (LPS) (n=25 and n=9 animals, respectively). The animals were allocated into treatment (anti-CD14) and control (IgG isotype or saline) groups. Inflammatory, hemostatic, physiological, and microbiological parameters were measured. The proinflammatory cytokines TNF-alpha, IL-1beta, IL-6, and IL-8, but not the anti-inflammatory cytokine IL-10, were efficiently inhibited by anti-CD14. Furthermore, anti-CD14 preserved the leukocyte count and significantly reduced granulocyte enzyme matrix metalloproteinase-9 release and expression of the granulocyte membrane activation molecule wCD11R3 (pig CD11b). The hemostatic markers thrombin-antithrombin III complexes and plasminogen activator inhibitor-1 were significantly attenuated. Anti-CD14 did not affect LPS or E. coli DNA levels. This study documents that CD14 inhibition efficiently attenuates the proinflammatory cytokine response and granulocyte activation and reverses the procoagulant state but does not interfere with LPS levels or bacterial counts in E. coli-induced sepsis.-Thorgersen, E. B., Hellerud, B. C., Nielsen, E. W., Barratt-Due, A., Fure, H., Lindstad, J. K., Pharo, A., Fosse, E., Tønnessen, T. I., Johansen, H. T., Castellheim, A., Mollnes, T. E. CD14 inhibition efficiently attenuates early inflammatory and hemostatic responses in Escherichia coli sepsis in pigs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Escherichia coli/pathogenicity , Lipopolysaccharide Receptors/immunology , Sepsis/drug therapy , Sepsis/immunology , Animals , Escherichia coli/genetics , Female , Flow Cytometry , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharides/toxicity , Male , Sepsis/chemically induced , Sepsis/microbiology , Swine , Tumor Necrosis Factor-alpha/metabolism
13.
Proc Natl Acad Sci U S A ; 106(37): 15861-6, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717455

ABSTRACT

Complement component C5 is crucial for experimental animal inflammatory tissue damage; however, its involvement in human inflammation is incompletely understood. The responses to gram-negative bacteria were here studied taking advantage of human genetic complement-deficiencies--nature's own knockouts--including a previously undescribed C5 defect. Such deficiencies provide a unique tool for investigating the biological role of proteins. The experimental conditions allowed cross-talk between the different inflammatory pathways using a whole blood model based on the anticoagulant lepirudin, which does not interfere with the complement system. Expression of tissue factor, cell adhesion molecules, and oxidative burst depended highly on C5, mediated through the activation product C5a, whereas granulocyte enzyme release relied mainly on C3 and was C5a-independent. Release of cytokines and chemokines was mediated to varying degrees by complement and CD14; for example, interleukin (IL)-1beta and IL-8 were more dependent on complement than IFN-gamma and IL-6, which were highly dependent on CD14. IL-1 receptor antagonist (IL-1ra) and IFN-gamma inducible protein 10 (IP-10) were fully dependent on CD14 and inversely regulated by complement, that is, complement deficiency and complement inhibition enhanced their release. Granulocyte responses were mainly complement-dependent, whereas monocyte responses were more dependent on CD14. Notably, all responses were abolished by combined neutralization of complement and CD14. The present study provides important insight into the comprehensive role of complement in human inflammatory responses to gram-negative bacteria.


Subject(s)
Complement System Proteins/deficiency , Complement System Proteins/genetics , Inflammation/genetics , Inflammation/immunology , Adolescent , Adult , Case-Control Studies , Cell Adhesion/immunology , Complement Activation , Complement C2/deficiency , Complement C2/genetics , Complement C5/deficiency , Complement C5/genetics , Escherichia coli/immunology , Female , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/pathogenicity , Humans , Immunity, Innate/genetics , In Vitro Techniques , Inflammation/etiology , Lipopolysaccharide Receptors/metabolism , Male , Models, Immunological , Monocytes/immunology , Monocytes/microbiology , Neisseria meningitidis/immunology , Phagocytosis , Respiratory Burst/immunology , Thromboplastin/biosynthesis
14.
Shock ; 32(3): 302-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19174740

ABSTRACT

The objective of this study was to establish a porcine analog of human meningococcal sepsis for pathophysiological investigations and possible future therapy in severe sepsis. Heat-killed Neisseria meningitidis was continuously infused in sublethal concentrations into 10 anesthetized 30-kg pigs (sepsis group). The dose was doubled every 30 min. Six pigs received saline only (control group). The changes described in the succeeding paragraphs were observed in the sepsis group but not in the control group. MAP was aimed to be kept normal by fluid infusion but declined after 3 h in parallel with a decrease in systemic vascular resistance. Pulmonary arterial pressure increased considerably after 30 to 45 min. A massive plasma extravasation was shown by increased hematocrit and a 50% reduction in plasma albumin content. Fluid accumulated in lungs, muscles, and jejunum, as shown by increased wet-dry ratios. Peak inspiratory pressures and fraction of inspired oxygen had to be increased. The cytokines TNF-alpha, IL-1beta, IL-6, IL-8, IL-10, and IL-12 increased markedly. Neutrophils fell to zero-levels, and platelets were markedly reduced. Thrombin-antithrombin complexes increased notably after 120 min. This is the first large animal model of sepsis using whole Neisseria meningitidis. The model simulates well central aspects of human meningococcal sepsis and could be used for future interventional studies.


Subject(s)
Meningococcal Infections/microbiology , Meningococcal Infections/pathology , Shock, Septic/microbiology , Shock, Septic/pathology , Animals , Disease Models, Animal , Female , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Meningococcal Infections/metabolism , Neisseria meningitidis/physiology , Random Allocation , Shock, Septic/metabolism , Swine , Tumor Necrosis Factor-alpha/metabolism
15.
Infect Immun ; 76(9): 4183-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18591229

ABSTRACT

The clinical presentation of meningococcal disease is closely related to the number of meningococci in the circulation. This study aimed to examine the activation of the innate immune system after being exposed to increasing and clinically relevant concentrations of meningococci. We incubated representative Neisseria meningitidis serogroup B (ST-32) and serogroup C (ST-11) strains and a lipopolysaccharide (LPS)-deficient mutant (the 44/76 lpxA mutant) in human serum and whole blood and measured complement activation and cytokine secretion and the effect of blocking these systems. HEK293 cells transfected with Toll-like receptors (TLRs) were examined for activation of NF-kappaB. The threshold for cytokine secretion and activation of NF-kappaB was 10(3) to 10(4) meningococci/ml. LPS was the sole inflammation-inducing molecule at concentrations up to 10(5) to 10(6) meningococci/ml. The activation was dependent on TLR4-MD2-CD14. Complement contributed to the inflammatory response at >or=10(5) to 10(6) meningococci/ml, and complement activation increased exponentially at >or=10(7) bacteria/ml. Non-LPS components initiated TLR2-mediated activation at >or=10(7) bacteria/ml. As the bacterial concentration exceeded 10(7)/ml, TLR4 and TLR2 were increasingly activated, independent of CD14. In this model mimicking human disease, the inflammatory response to N. meningitidis was closely associated with the bacterial concentration. Therapeutically, CD14 inhibition alone was most efficient at a low bacterial concentration, whereas addition of a complement inhibitor may be beneficial when the bacterial load increases.


Subject(s)
Complement System Proteins/immunology , Neisseria meningitidis/growth & development , Sepsis/immunology , Sepsis/microbiology , Toll-Like Receptors/immunology , Cell Line , Complement Activation , Cytokines/metabolism , Humans , Lipopolysaccharides/biosynthesis , NF-kappa B/metabolism
16.
Tidsskr Nor Laegeforen ; 125(15): 2031-2, 2005 Aug 11.
Article in Norwegian | MEDLINE | ID: mdl-16114124

ABSTRACT

Inhaled corticosteroids are a well established and effective treatment for asthma in children. However, some children develop systemic side effects including adrenal suppression when using moderate to high doses. Over the last few years, several severe acute adrenal crises with hypoglycaemia in patients using inhaled corticosteroids have been reported. Normally these patients do not develop a Cushingoid appearance and their height is not necessarily affected. We present a three-years-old boy that was unconscious at admittance. From the age of 6 months he had had asthma, treated with fluticasone propionate. The last year his asthma had been difficult to control, and he was given 750-1000 g/day in combination with salmeterol and a leucotriene antagonist. The day before admittance he had been ill with fever, had poor intake of food, and no intake of his regular medication. He was found unconscious in the morning. At admittance the blood glucose was 1.8. His cortisol axis was partially suppressed, probably as a result of the high doses of fluticasone propionate that had been administered. When treating asthmatic children it is important to use the lowest possible dose of inhaled corticosteroids. Those in need of higher doses should be carefully followed up with respect to systemic side effects. In emergency situations, systemic steroids should be used liberally in these children.


Subject(s)
Albuterol/analogs & derivatives , Androstadienes/adverse effects , Asthma/drug therapy , Bronchodilator Agents/adverse effects , Hypoglycemia/chemically induced , Administration, Inhalation , Albuterol/administration & dosage , Albuterol/adverse effects , Androstadienes/administration & dosage , Bronchodilator Agents/administration & dosage , Child, Preschool , Drug Therapy, Combination , Fluticasone , Humans , Male , Salmeterol Xinafoate
SELECTION OF CITATIONS
SEARCH DETAIL
...