Skip to main content
Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases 8/2019

Open Access 14.05.2019 | Original Article

Plasma concentrations of secretory leukocyte protease inhibitor (SLPI) differ depending on etiology and severity in community-onset bloodstream infection

verfasst von: Anna Lange, Sara Cajander, Anders Magnuson, Jonas Sundén-Cullberg, Kristoffer Strålin, Olof Hultgren

Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases | Ausgabe 8/2019

Abstract

The severity of bloodstream infections (BSI) depends on pathogen, source, and host factors. Secretory leukocyte protease inhibitor (SLPI) counteracts tissue damage, balances inflammation, and is increased in pneumonia and sepsis. We aimed to evaluate whether SLPI production differs depending on etiology, disease severity, and sex in BSI and to correlate SLPI with markers of inflammation and immunosuppression. Of the adult patients with BSI, 109 were included and sampled repeatedly, from hospital admission through day 28. Controls (blood donors) were sampled twice. SLPI in plasma was measured with enzyme-linked immunosorbent assay (ELISA) technique. Streptococcus pneumoniae and Staphylococcus aureus etiology were associated with higher SLPI than Escherichia coli on days 1–2 and 3. On day 1–2, subjects with sepsis had higher SLPI concentrations than those with non-septic BSI. Pneumonia was associated with higher SLPI than a non-pulmonary source of infection. SLPI co-varied with inflammatory markers. SLPI concentrations did not differ with regard to sex in the full cohort, but men with pneumonia had higher SLPI than women on day 1–2. S. pneumoniae and S. aureus BSI were associated with higher SLPI, when compared to E. coli. Severity and pneumonia, as well as male sex in the pneumonia sub-cohort, were factors independently associated with higher SLPI.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1007/​s10096-019-03567-2) contains supplementary material, which is available to authorized users.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The clinical course of a bacterial infection is determined by the interaction of host factors, the infected organ, and the pathogen. [1]. Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection, but there are currently no criteria for measuring the dysregulated immune response, which can manifest as disproportionate pro-inflammation and/or a state of immunosuppression [2]. The risk of death and morbidity in sepsis is still considerable [3, 4]. The causative microorganism is identified in 50–60%, and in 20–30% there is an associated bloodstream infection (BSI), defined as growth of one or more bacterial or fungal pathogens in one or more blood cultures [1, 57]. The reported incidence of BSI is in average 140–160/100,000/year in high-income countries, and the three most common etiologies are Escherichia coli, Staphylococcus aureus, and Streptococcus pneumoniae [8].
Secretory leukocyte protease inhibitor (SLPI) is a protein that has come to be seen as an important regulator of inflammation [9]. SLPI was first isolated in pulmonary secretions, and has been recognized as being not only a tissue protector that inhibits neutrophil-derived proteases but also for having other immunological tasks [10]. Similarly to antimicrobial peptides, SLPI exhibits antibacterial, antifungal, and antiviral properties, and it is known to balance pro-inflammation by downregulating the NFκB pathway [1116]. In addition, recent research suggests that SLPI may further modulate immunity by regulating neutrophil maturation, and through inhibition of lymphocyte proliferation and the formation of neutrophil extracellular traps [1719].
SLPI is primarily of epithelial cell origin, but is also formed by dendritic cells, macrophages, and neutrophils [11, 2022]. The production and secretion of SLPI is regulated by pro-inflammatory stimuli [21, 23, 24].
The biological role of SLPI expression in sepsis remains only partially understood, but experimental evidence suggests that SLPI protects from detrimental inflammation. In humans, plasma SLPI has been found to be increased in sepsis, and to be associated with the degree of organ dysfunction [24].
Studies investigating SLPI in the context of sepsis are limited, and SLPI production over the course of BSI, or in relation to clinical characteristics and etiology has not been studied. We hypothesized that SLPI expression might differ depending on bacterial etiology and the source of infection. Thus, we aimed to study SLPI in a cohort of well-characterized patients with BSI followed 4 weeks. With previous findings of higher plasma SLPI concentrations in community-acquired pneumonia (CAP) in males, we also intended to study SLPI in relation to sex [25]. Finally, we wanted to see if SLPI correlates to markers of inflammation/immunosuppression.

Methods

Setting and study population

A prospective study of patients with BSI was conducted at Örebro University Hospital, Örebro, Sweden, between 2011 and 2014. Patients > 18 years, admitted to the Departments of Infectious Diseases and Internal Medicine, with a suspected infection, and in whom a blood culture drawn on hospital admission (day 0) showed growth of clinically significant bacteria within 3 days, were eligible for inclusion. Exclusion criteria were infection with HIV, hepatitis B and C, or previous inclusion in the study.
Blood samples were drawn from study subjects on day(s) 0, 1–2, 3, 7 ± 1, 14 ± 2, and 28 ± 4. HLA-DR expression on monocytes was measured from day 1–2. HLA-DR data is elsewhere described in detail [26]. CRP, neutrophil count, and lymphocyte count were analyzed with accredited routine laboratory methods. Patient data was obtained from medical records. Blood and plasma donors (n = 31, not sex- and age-matched) served as controls, and were sampled twice, 4 weeks apart. EDTA plasma was kept at − 80 °C pending analysis.

Blood and other cultures

Two blood cultures, each consisting of 20 mL blood distributed equally between one aerobic and one anaerobic bottle, were incubated in a Bactec blood culturing system (Becton Dickinson, Franklin Lakes, NJ, USA). The bacterial species were determined by routine laboratory diagnostic procedures. Other cultures were taken depending on clinical suspicion of diagnosis according to clinical routine, and before administration of antibiotics when possible.

Definitions

Sepsis was defined as an increase of the Sequential Organ Failure Assessment (SOFA) score by ≥ 2 points from baseline, according to Sepsis-3 definitions [2].
The diagnosis of pneumonia required a radiographic pulmonary infiltrate. Urinary tract infection (UTI) diagnosis required a urine culture testing positive for the same bacteria as the one detected in blood, and infective endocarditis was diagnosed based on the revised Duke criteria.

ELISA analyses

SLPI in EDTA plasma was analyzed in duplicate at 1:100 dilution with Human SLPI ELISA Kit, Hycult Biotech, the Netherlands, detection range 78–5000 pg/mL, according to the manufacturer’s instructions.

Statistics

IBM SPSS Statistics for Windows, version 24.0 (IBM Corp., Armonk, NY, USA) and STATA release 14 (StataCorp LP, College Station, TX, USA) were used. Due to non-normal distribution of SLPI, analyses were performed on log10 scale, or with non-parametric methods.
Patients were classified according to bacterial etiology (E. coli/S. aureus/S. pneumonia/other), source (pneumonia/other), and severity (sepsis/non-septic BSI). Age, sex, duration of illness, Charlson score, and severity were compared between etiology groups with chi-square, Fisher, unpaired t, or Mann-Whitney test, according to variable distribution.
Differences in SLPI on day 1–2 were evaluated with linear regression. The strategy was (1) unadjusted analyses of (a) bacterial etiology, (b) pneumonia/other sources, and (c) sepsis/no sepsis, in the full BSI cohort, and (d) sex in the pneumonia sub-cohort and (2) adjusted models: (a) and (b) were adjusted for age, sex, and SOFA score increase on admission, (c) for age and sex, and (d) for age and SOFA score increase.
The above analyses were performed on complete cases (n = 97), and, to reduce potential bias due to missing data, on all patients (n = 109) after multiple imputation (MI), based on Rubin’s concept [27]. Age, sex, duration of illness, SOFA score increase, bacterial etiology, immunosuppression, and Charlson score 0, 1, 2, or more were used as predictors for the SLPI imputation.
Linear mixed model with random intercept was used to evaluate mean differences of SLPI over days 1–2 to 28, for E. coli, S. aureus, and S. pneumoniae. Fixed factors were day, etiology, and their interaction term. The mean differences of SLPI between etiology groups on each time point were compared, and p values were Bonferroni-corrected. Since mixed model assumes that missing data are missing at random, demographic and clinical characteristics were compared with basic statistical methods as described above, between subjects with full and incomplete SLPI series.
Unadjusted and age- and sex-adjusted linear regression was performed to compare SLPI in BSI and controls, day 0 and 28.
Spearman correlation (rs) was used to correlate SLPI and inflammatory markers on day 1–2 and 7.
A p value < 0.05 was considered statistically significant.

Results

Characteristics of the study population

One-hundred-sixteen patients were enrolled. Seven were excluded due to growth of non-pathogenic bacteria (n = 5), or no available plasma samples (n = 2), leaving 109 valid subjects. Plasma was available from 46 subjects on admission, 97 (day 1–2), 68 (day 3), 86 (day 7), 78 (day 14), and 72 (day 28). Subjects with incomplete plasma series were older than those with full series (p = 0.02), but not otherwise significantly differing in baseline characteristics (data not shown).
Baseline and clinical characteristics and etiology are detailed in Table 1. The main sources of infection were pneumonia, UTI, skin/soft tissue, and joint infection. Fifty-four subjects had sepsis. Subjects with S. pneumonia BSI had higher SOFA score increases and frequency of sepsis when compared to E. coli. Ninety-day mortality was 10%. Eighty-nine percent of the study subjects received correct antibiotic treatment within 6 h. One subject had growth of ESBL-producing E. coli, and none had MRSA. Two had neutropenic sepsis (< 1000 neutrophils/mm3), one with polymicrobial etiology, one with beta-hemolytic streptococcal BSI.
Table 1
Demographic and clinical patient characteristics
Bloodstream infection etiology
Characteristics
HC (n = 31)
E. coli (n = 25)
S. aureus (n = 27)
S. pneumoniae (n = 29)
Othera (n = 28)
Age mean (SD)
52 (8)
71 (18)
72 (17)
68 (11)
68 (12)
Sex maled
24 (77.4)
11 (44.0)
23 (85.2)
9 (31.0)
16 (57.1)*
Number of days of diseased
 
2 (1–3)
3.5 (2–4)
2 (2–4)
1.5 (0–4)*
SOFA increase 0d
 
11 (44.0)
8 (29.6)
1 (3.4)
6 (21.4)*
SOFA increase 1d
 
8 (32.0)
6 (22.2)
7 (24.1)
8 (28.6)*
SOFA increase 2d
 
3 (12)
6 (22.2)
9 (31.0)
4 (14.3)*
SOFA increase ≥ 3d
 
3 (12)
7 (25.9)
12 41.4)
10 (35.7)*
Intensive care admission
 
2 (8.0)
3 (11.1)
6 (20.7)
3 (10.7)
Sepsis
 
6 (24.0)
13 (48)
21 (72.4)
14 (50)
Comorbid conditions
  Immunosuppressionb
 
4 (16.0)
1 (3.7)
3 (10.3)
3 (10.7)
  Congestive heart failure
 
4 (16.0)
5 (18.5)
3 (10.3)
4 (14.3)
  Peripheral vascular disease
 
1 (4.0)
1 (3.7)
1 (3.4)
2 (7.1)
  Moderate or severe kidney disease
 
3 (12.0)
3 (11.1)
0
2 (7.1)
  Chronic liver disease
 
0
0
0
0
  Dementia
 
2 (8.0)
1 (3.7)
0
2 (7.1)
  Connective tissue disease
 
3 (12.0)
1 (3.7)
4 (13.8)
1 (3.6)
  Any tumor within the last 5 years
 
2 (8.0)
1 (3.7)
0
2 (7.1)
  Ischemic heart disease
 
8 (32.0)
7 (25.9)
4 (13.8)
3 (10.7)
  Chronic lung disease
 
0
1 (3.7)
5 (17.2)
2 (7.1)
  Diabetes with complications
 
7 (28.0)
0
0
7 (25.0)
  Charlson comorbidity score
 
2 (0–7)
1 (0–8)
1 (0–8)
1 (0–5)
Primary focus of infection
  Lung
  
1 (3.7)
23 (79.3)
3 (10.7)
  Airways
   
2 (6.9)
 
  Urinary tract
 
20 (80.0)
  
7 (25.0)
  Skin/soft tissue
  
5 (18.5)
 
5 (17.9)
  Arthritis/osteomyelitis
  
10 (37.0)
1 (3.4)
 
  Abdominal
 
1 (4.0)
  
6 (21.4)
  Endocarditis
 
5 (18.5)
  
3 (10.7)
  Meningitis
   
3 (10.3)
 
  Otherc
 
1 (4.0)
2 (7.4)
 
1 (3.6)
  Unknown
 
3 (12.0)
4 (14.8)
 
3 (10.7)
aBetahemolytic streptocci, n = 7; Klebsiella pneumoniae, n = 5,; Polymicrobial, n = 5; Enterococci, n = 4; Pseudomonas, n = 2; Salmonella, n = 2; Streptococcus anginosus, n = 1; Haemophilus influenzae, n = 1; Actinobacillum schalii, n = 1
bPrednisolone > 20-mg daily dose, chemotherapy, or neutropenia prior to sepsis
cStent graft infection, n = 2; primary bloodstream infection, n = 2. Data is presented as n (%) unless otherwise stated. # median (range)
dSignificant etiology group differences. Sex: E. coli-S. aureus, p = 0.02; S. aureus-S. pneumoniae < 0.01. Number of days of disease: E. coli-S. aureus, p < 0.01. SOFA score increase on admission: E. coli-S. pneumoniae, p < 0.01

SLPI and etiology: bacteria groups and source of infection

SLPI concentrations for bacterial subgroups on day 1–2 are shown in Fig. 1a. Linear regression showed a statistically significantly higher unadjusted mean SLPI concentration in S. pneumoniae and S. aureus etiology, when compared to E. coli in the unadjusted and adjusted (age, sex, and initial SOFA score increase) models (Table 2). Analysis with MI gave the same statistically significant findings for S. pneumoniae. For S. aureus it showed B = 0.12, 95% CI 0.02–0.23, p = 0.02 in the unadjusted, and B = 0.08, 95% CI − 0.02–0.18, p = 0.1 in the adjusted model.
Table 2
Linear regression with log10SLPI on day 1–2 (n = 97) as outcome variable
 
n (%)
Unadjusted
Adjusted for age, sex, and SOFA on admission
B
95% CI
p
B
95% CI
p
Age, per year
 
0.002
− 0.001 to 0.005
0.12
0.0009
− 0.001 to 0.003
0.43
Female sex
44 (45)
ref
ref
 
ref
ref
 
Male sex
53 (55)
0.01
− 0.06 to 0.09
0.73
0.03
− 0.04 to 0.09
0.43
SOFA increase, per unit
 
0.046
0.029 to 0.062
< 0.01
0.039
0.022 to 0.056
< 0.01
E. coli
20 (21)
ref
ref
 
ref
ref
 
S. aureus
24 (25)
0.15
0.06 to 0.25
< 0.01
0.10
0.008 to 0.20
0.04
S. pneumoniae
27 (28)
0.20
0.10 to 0.30
< 0.01
0.14
0.05 to 0.23
< 0.01
Other etiologies
26 (27)
0.07
− 0.02 to 0.16
0.17
0.02
− 0.07 to 0.11
0.65
Subjects with pneumonia had higher mean SLPI concentrations on day 1–2 than other sources of infection: unadjusted (B = 0.13, 95% CI 0.05–0.21, p < 0.01) and adjusted (B = 0.10, 95% CI 0.02–0.17, p = 0.01) (Fig. 1b). The same conclusion was made with MI.

SLPI and disease severity

The initial SOFA score increase was positively associated with SLPI concentrations, as shown in Table 2. Subjects with sepsis had higher mean SLPI on day 1–2 when compared to non-septic BSI: unadjusted (B = 0.17, 95% CI 0.10–0.23, p < 0.01) and adjusted (age and sex) (B = 0.16, 95% CI 0.10–0.23, p < 0.01) (Fig. 2). Analysis using MI produced similar results.

SLPI and sex

The male sex was not associated with higher SLPI in controls (B = − 0.001, 95% CI − 0.12–0.12, p = 0.98) or on day 1–2 in the full BSI cohort (B = − 0.05, 95% CI − 0.14–0.03, p = 0.2). In the pneumonia sub-cohort (men n = 9, women n = 17) however, the male sex showed statistically significantly higher mean SLPI, unadjusted (B = 0.11, 95% CI 0.01–0.20, p = 0.03), and adjusted (age and SOFA score increase) (B = 0.11, 95% CI 0.03–0.19, p = 0.01) (Fig. 3).

SLPI dynamics

SLPI concentrations for bacterial etiology groups on days 0–28 are shown in Fig. 1. The linear mixed model interaction test between bacterial etiology and time was not significant (p = 0.29), showing no statistically significant different overall mean response over time. However, S. pneumonia etiology had higher mean SLPI concentration on day 3 (p = 0.01) when compared to E. coli, but not on day 7 (p = 0.06) and thereafter. S. aureus etiology had higher mean SLPI compared to E. coli, on day 3 (p = 0.02), day 14 (p = 0.03), and day 28 (p = 0.04), but not on day 7 (p = 0.36).

SLPI in BSI and controls

Subjects with BSI had significantly higher SLPI concentrations on day 1–2 when compared to controls on day 0: unadjusted (B = 0.36, 95% CI 0.29–0.43, p < 0.01) and adjusted (age and sex) (B = 0.33, 95% CI 0.25–0.42, p < 0.01). BSI was associated with higher SLPI on day 28 in both unadjusted (B = 0.14, 95% CI 0.08–0.20, p < 0.01) and adjusted analysis (B = 0.10, 95% CI 0.03–0.17, p < 0.01) (Fig. 2).

SLPI and other biomarkers

SLPI did not correlate to lactate on hospital admission (rs = 0.27, p = 0.3). SLPI was significantly positively correlated to CRP and neutrophil count, on days 1–2 and 7. It did not correlate to lymphocyte count on day 1–2 but a negative significant correlation was present on day 7. SLPI correlated negatively to HLA-DR on days 1–2 and 7 (Fig. 4a–h).

Discussion

This study reports independent associations between bacterial etiology, disease severity, lung focus, and plasma concentration of SLPI in community-onset BSI. This is, to our knowledge, the first clinical study of SLPI as it relates to microbial etiology.
We found that, on days 1–2 and 3 after admission, subjects with S. pneumoniae and S. aureus etiology had higher plasma SLPI than E. coli. With account taken for the potential bias of missing samples over time, we also saw higher SLPI in S. aureus etiology compared to E. coli later in the studied period. Despite known differences in bacterial virulence, tissue tropism, and pathogen sensing by the immune system, sepsis studies rarely account for the causative pathogen [28]. We recently published another study based on this patient cohort, showing that mHLA-DR expression varies according to bacterial etiology, with low initial mHLA-DR in S. aureus and S. pneumoniae BSI [26]. A few studies have compared inflammatory markers in gram-positive and gram-negative BSI. Results are conflicting, perhaps due to study group variations. A study of general ICU patients found higher IL-6 and CRP in gram-negative sepsis [29]. Another group looked at abdominal sepsis, reporting higher levels of most pro- (TNF-α, IFN-γ, and CXCL8) and antiinflammatory (IL-1ra, IL-4, and IL-10) mediators measured, in pure gram-negative etiology. Contrastingly, a study of patients with “gram-positive cocci” (mainly S. aureus) and “gram-negative bacilli” (mainly E. coli) found no difference in IL-6 and CXCL8 concentrations [30, 31].
SLPI concentrations were associated with initial SOFA score increase, and were higher in early septic, compared to uncomplicated BSI, supporting previous results from human and animal studies. One study found an association between SLPI levels in human sepsis and the degree of organ failure [24]. Another reported increased LPS-induced immune cell activation in SLPI−/− mice, more severe disease, and higher mortality in sepsis-challenged knockout mice compared to wild type (WT), and a third study correspondingly found increased LPS-induced cytokine secretion in lymph nodes in SLPI−/− mice when compared to WT [22, 32].
Pulmonary sepsis is associated with high mortality [33]. We found that pneumonia was associated with higher SOFA score on admission (p < 0.01), than other etiologies, but adjusted analyses showed an independent association between pneumonia and SLPI, which concurs with previous reports by us and others [25, 34]. S. pneumoniae is the predominant pathogen in CAP, and made out 85% of BSI in our pneumonia sub-cohort. No patient had E. coli, and only one had S. aureus, which ruled out etiology-stratified analysis. SLPI is produced by epithelial cells and macrophages in the lungs, and pulmonary secretion concentrations of SLPI are increased in pneumonia [35, 36]. One study reported that, in pneumonia, SLPI in plasma was proportional to the extent of lung tissue involved, suggesting SLPI spill-over into the circulation [34]. SLPI release from specific granules upon neutrophil activation might also contribute to increased plasma SLPI [37]. Additionally, SLPI is secreted by other epithelial cells, and another source of plasma SLPI could be protein leakage from other epithelial sites than the lungs [9]. A study of acetaminophen-induced acute liver failure exemplified this, showing that circulating SLPI derived from the liver [38]. It is yet possible that epithelial affection is more significant in pneumonia, explaining some of the observed differences.
Despite the low number of study subjects, and in line with our previous findings with a larger cohort of CAP, we report higher plasma SLPI, independently of severity, in men than in women with pneumonia and BSI on day 1–2 [25]. Due to loss of sampling over time, we could not assess the dynamics of this sex-related difference. Through unknown mechanisms, the sexes display differences in infection and immunity already in childhood. Females are predisposed to autoimmunity and better vaccine responses, whereas males are more susceptible to some infections [39, 40]. In terms of sepsis, a male preponderance has been demonstrated in both adults and children, and as to pneumonia, male sex is a risk factor for severity [1, 7, 41, 42]. Experimental studies indicate that female sex hormones are protective in sepsis [43]. Interestingly, sex hormones, or sex hormone receptors, have been suggested to be involved in the regulation of SLPI expression [4446].
SLPI covaried with CRP and neutrophil count, and remained elevated when compared to controls throughout this study. The biological significance of circulating SLPI is unknown, but SLPI was detected in the nucleus of peripheral blood monocytes in sepsis patients, and it was shown in vitro that monocytes and B cells internalize exogenous SLPI [16, 47]. SLPI does not influence mHLA-DR expression in vitro, but the observed inverse correlation between these two markers might suggest that SLPI in plasma does not only reflect a pro-inflammatory state or tissue damage, but is possibly linked to sepsis-induced immunosuppression [38]. Future studies might identify whether circulating SLPI penetrates immune cells to antagonize inflammatory responses, and if the correlation between SLPI and disease severity, as demonstrated by us and others, relates to that.
This study has some limitations, including the limited number of subjects, half of them not sampled for plasma on admission, making the study underpowered for etiological comparison before antibiotic were given. Secondly, missing data may affect the validity of the over-time analyses, and equivalently, MI analysis showed lower association for S. aureus etiology; hence, these results are interpreted with caution. Furthermore, Sweden has a low degree of antibiotic resistance compared to most countries, and the greater part of subjects with E. coli BSI had a UTI, which is known to be associated with milder disease than an abdominal focus [48]. Therefore, in other settings, conclusions might have been different.
Strengths of this study include clear definitions of BSI and etiology, and varying disease severity, reflecting the heterogeneity of BSI and sepsis. Development of treatments targeting the dysregulated host response in sepsis will require deeper knowledge in how host-related factors, e.g., sex, and specific pathogens, influence the immune response.
In summary, this study shows differences in SLPI blood concentrations related to bacterial etiology, disease severity, and source in BSI, plus a sex-related divergence in SLPI concentrations in pneumonia. SLPI is noticed for versatile immunological functions, and our results warrant further studies that elucidate the role of SLPI in severe bacterial infections.

Acknowledgments

We thank patients and controls for their participation, and Ulla Larsson, biomedical technician, for her excellent laboratory assistance.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

Ethical approval was achieved from the Regional Ethics Review Board of Uppsala, Sweden (02/25/2009, file number 2009/024), and the study was conducted in accordance with the Declaration of Helsinki.
Written informed consent was obtained from the patient or relative.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Vincent JL, Sakr Y, Sprung CL, Ranieri VM, Reinhart K, Gerlach H, Moreno R, Carlet J, Le Gall JR, Payen D (2006) Sepsis in European intensive care units: results of the SOAP study. Crit Care Med 34(2):344–353PubMed Vincent JL, Sakr Y, Sprung CL, Ranieri VM, Reinhart K, Gerlach H, Moreno R, Carlet J, Le Gall JR, Payen D (2006) Sepsis in European intensive care units: results of the SOAP study. Crit Care Med 34(2):344–353PubMed
2.
Zurück zum Zitat Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, Hotchkiss RS, Levy MM, Marshall JC, Martin GS, Opal SM, Rubenfeld GD, van der Poll T, Vincent JL, Angus DC (2016) The third international consensus definitions for Sepsis and septic shock (Sepsis-3). JAMA 315(8):801–810PubMedPubMedCentral Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, Hotchkiss RS, Levy MM, Marshall JC, Martin GS, Opal SM, Rubenfeld GD, van der Poll T, Vincent JL, Angus DC (2016) The third international consensus definitions for Sepsis and septic shock (Sepsis-3). JAMA 315(8):801–810PubMedPubMedCentral
3.
Zurück zum Zitat Stevenson EK, Rubenstein AR, Radin GT, Wiener RS, Walkey AJ (2014) Two decades of mortality trends among patients with severe sepsis: a comparative meta-analysis*. Crit Care Med 42(3):625–631PubMedPubMedCentral Stevenson EK, Rubenstein AR, Radin GT, Wiener RS, Walkey AJ (2014) Two decades of mortality trends among patients with severe sepsis: a comparative meta-analysis*. Crit Care Med 42(3):625–631PubMedPubMedCentral
4.
5.
Zurück zum Zitat Martin GS, Mannino DM, Eaton S, Moss M (2003) The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med 348(16):1546–1554 Martin GS, Mannino DM, Eaton S, Moss M (2003) The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med 348(16):1546–1554
6.
Zurück zum Zitat Phua J, Ngerng W, See K, Tay C, Kiong T, Lim H, Chew M, Yip H, Tan A, Khalizah H, Capistrano R, Lee K, Mukhopadhyay A (2013) Characteristics and outcomes of culture-negative versus culture-positive severe sepsis. Crit Care 17(5):R202PubMedPubMedCentral Phua J, Ngerng W, See K, Tay C, Kiong T, Lim H, Chew M, Yip H, Tan A, Khalizah H, Capistrano R, Lee K, Mukhopadhyay A (2013) Characteristics and outcomes of culture-negative versus culture-positive severe sepsis. Crit Care 17(5):R202PubMedPubMedCentral
7.
Zurück zum Zitat Esper AM, Moss M, Lewis CA, Nisbet R, Mannino DM, Martin GS (2006) The role of infection and comorbidity: factors that influence disparities in sepsis. Crit Care Med 34(10):2576–2582PubMedPubMedCentral Esper AM, Moss M, Lewis CA, Nisbet R, Mannino DM, Martin GS (2006) The role of infection and comorbidity: factors that influence disparities in sepsis. Crit Care Med 34(10):2576–2582PubMedPubMedCentral
8.
Zurück zum Zitat Laupland KB (2013) Incidence of bloodstream infection: a review of population-based studies. Clin Microbiol Infect 19(6):492–500PubMed Laupland KB (2013) Incidence of bloodstream infection: a review of population-based studies. Clin Microbiol Infect 19(6):492–500PubMed
9.
Zurück zum Zitat Majchrzak-Gorecka M, Majewski P, Grygier B, Murzyn K, Cichy J (2016) Secretory leukocyte protease inhibitor (SLPI), a multifunctional protein in the host defense response. Cytokine Growth Factor Rev 28:79–93PubMed Majchrzak-Gorecka M, Majewski P, Grygier B, Murzyn K, Cichy J (2016) Secretory leukocyte protease inhibitor (SLPI), a multifunctional protein in the host defense response. Cytokine Growth Factor Rev 28:79–93PubMed
10.
Zurück zum Zitat Thompson RC, Ohlsson K (1986) Isolation, properties, and complete amino acid sequence of human secretory leukocyte protease inhibitor, a potent inhibitor of leukocyte elastase. Proc Natl Acad Sci U S A 83(18):6692–6696PubMedPubMedCentral Thompson RC, Ohlsson K (1986) Isolation, properties, and complete amino acid sequence of human secretory leukocyte protease inhibitor, a potent inhibitor of leukocyte elastase. Proc Natl Acad Sci U S A 83(18):6692–6696PubMedPubMedCentral
11.
Zurück zum Zitat Hiemstra PS, Maassen RJ, Stolk J, Heinzel-Wieland R, Steffens GJ, Dijkman JH (1996) Antibacterial activity of antileukoprotease. Infect Immun 64(11):4520–4524PubMedPubMedCentral Hiemstra PS, Maassen RJ, Stolk J, Heinzel-Wieland R, Steffens GJ, Dijkman JH (1996) Antibacterial activity of antileukoprotease. Infect Immun 64(11):4520–4524PubMedPubMedCentral
12.
Zurück zum Zitat Gomez SA, Arguelles CL, Guerrieri D, Tateosian NL, Amiano NO, Slimovich R, Maffia PC, Abbate E, Musella RM, Garcia VE, Chuluyan HE (2009) Secretory leukocyte protease inhibitor: a secreted pattern recognition receptor for mycobacteria. Am J Respir Crit Care Med 179(3):247–253PubMed Gomez SA, Arguelles CL, Guerrieri D, Tateosian NL, Amiano NO, Slimovich R, Maffia PC, Abbate E, Musella RM, Garcia VE, Chuluyan HE (2009) Secretory leukocyte protease inhibitor: a secreted pattern recognition receptor for mycobacteria. Am J Respir Crit Care Med 179(3):247–253PubMed
13.
Zurück zum Zitat Tomee JF, Hiemstra PS, Heinzel-Wieland R, Kauffman HF (1997) Antileukoprotease: an endogenous protein in the innate mucosal defense against fungi. J Infect Dis 176(3):740–747PubMed Tomee JF, Hiemstra PS, Heinzel-Wieland R, Kauffman HF (1997) Antileukoprotease: an endogenous protein in the innate mucosal defense against fungi. J Infect Dis 176(3):740–747PubMed
14.
Zurück zum Zitat McNeely TB, Dealy M, Dripps DJ, Orenstein JM, Eisenberg SP, Wahl SM (1995) Secretory leukocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency virus 1 activity in vitro. J Clin Invest 96(1):456–464PubMedPubMedCentral McNeely TB, Dealy M, Dripps DJ, Orenstein JM, Eisenberg SP, Wahl SM (1995) Secretory leukocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency virus 1 activity in vitro. J Clin Invest 96(1):456–464PubMedPubMedCentral
15.
Zurück zum Zitat Taggart CC, Cryan SA, Weldon S, Gibbons A, Greene CM, Kelly E, Low TB, O'Neill SJ, McElvaney NG (2005) Secretory leucoprotease inhibitor binds to NF-kappaB binding sites in monocytes and inhibits p65 binding. J Exp Med 202(12):1659–1668PubMedPubMedCentral Taggart CC, Cryan SA, Weldon S, Gibbons A, Greene CM, Kelly E, Low TB, O'Neill SJ, McElvaney NG (2005) Secretory leucoprotease inhibitor binds to NF-kappaB binding sites in monocytes and inhibits p65 binding. J Exp Med 202(12):1659–1668PubMedPubMedCentral
16.
Zurück zum Zitat Taggart CC, Greene CM, McElvaney NG, O'Neill S (2002) Secretory leucoprotease inhibitor prevents lipopolysaccharide-induced IkappaBalpha degradation without affecting phosphorylation or ubiquitination. J Biol Chem 277(37):33648–33653PubMed Taggart CC, Greene CM, McElvaney NG, O'Neill S (2002) Secretory leucoprotease inhibitor prevents lipopolysaccharide-induced IkappaBalpha degradation without affecting phosphorylation or ubiquitination. J Biol Chem 277(37):33648–33653PubMed
17.
Zurück zum Zitat Guerrieri D, Tateosian NL, Maffia PC, Reiteri RM, Amiano NO, Costa MJ, Villalonga X, Sanchez ML, Estein SM, Garcia VE, Sallenave JM, Chuluyan HE (2011) Serine leucocyte proteinase inhibitor-treated monocyte inhibits human CD4(+) lymphocyte proliferation. Immunology 133(4):434–441PubMedPubMedCentral Guerrieri D, Tateosian NL, Maffia PC, Reiteri RM, Amiano NO, Costa MJ, Villalonga X, Sanchez ML, Estein SM, Garcia VE, Sallenave JM, Chuluyan HE (2011) Serine leucocyte proteinase inhibitor-treated monocyte inhibits human CD4(+) lymphocyte proliferation. Immunology 133(4):434–441PubMedPubMedCentral
18.
Zurück zum Zitat Klimenkova O, Ellerbeck W, Klimiankou M, Unalan M, Kandabarau S, Gigina A, Hussein K, Zeidler C, Welte K, Skokowa J (2014) A lack of secretory leukocyte protease inhibitor (SLPI) causes defects in granulocytic differentiation. Blood 123(8):1239–1249PubMed Klimenkova O, Ellerbeck W, Klimiankou M, Unalan M, Kandabarau S, Gigina A, Hussein K, Zeidler C, Welte K, Skokowa J (2014) A lack of secretory leukocyte protease inhibitor (SLPI) causes defects in granulocytic differentiation. Blood 123(8):1239–1249PubMed
19.
Zurück zum Zitat Zabieglo K, Majewski P, Majchrzak-Gorecka M, Wlodarczyk A, Grygier B, Zegar A, Kapinska-Mrowiecka M, Naskalska A, Pyrc K, Dubin A, Wahl SM, Cichy J (2015) The inhibitory effect of secretory leukocyte protease inhibitor (SLPI) on formation of neutrophil extracellular traps. J Leukoc Biol 98(1):99–106PubMedPubMedCentral Zabieglo K, Majewski P, Majchrzak-Gorecka M, Wlodarczyk A, Grygier B, Zegar A, Kapinska-Mrowiecka M, Naskalska A, Pyrc K, Dubin A, Wahl SM, Cichy J (2015) The inhibitory effect of secretory leukocyte protease inhibitor (SLPI) on formation of neutrophil extracellular traps. J Leukoc Biol 98(1):99–106PubMedPubMedCentral
20.
Zurück zum Zitat Sallenave JM, Si Tahar M, Cox G, Chignard M, Gauldie J (1997) Secretory leukocyte proteinase inhibitor is a major leukocyte elastase inhibitor in human neutrophils. J Leukoc Biol 61(6):695–702PubMed Sallenave JM, Si Tahar M, Cox G, Chignard M, Gauldie J (1997) Secretory leukocyte proteinase inhibitor is a major leukocyte elastase inhibitor in human neutrophils. J Leukoc Biol 61(6):695–702PubMed
21.
Zurück zum Zitat Jin F, Nathan CF, Radzioch D, Ding A (1998) Lipopolysaccharide-related stimuli induce expression of the secretory leukocyte protease inhibitor, a macrophage-derived lipopolysaccharide inhibitor. Infect Immun 66(6):2447–2452PubMedPubMedCentral Jin F, Nathan CF, Radzioch D, Ding A (1998) Lipopolysaccharide-related stimuli induce expression of the secretory leukocyte protease inhibitor, a macrophage-derived lipopolysaccharide inhibitor. Infect Immun 66(6):2447–2452PubMedPubMedCentral
22.
Zurück zum Zitat Samsom JN, van der Marel AP, van Berkel LA, van Helvoort JM, Simons-Oosterhuis Y, Jansen W, Greuter M, Nelissen RL, Meeuwisse CM, Nieuwenhuis EE, Mebius RE, Kraal G (2007) Secretory leukoprotease inhibitor in mucosal lymph node dendritic cells regulates the threshold for mucosal tolerance. J Immunol 179(10):6588–6595PubMed Samsom JN, van der Marel AP, van Berkel LA, van Helvoort JM, Simons-Oosterhuis Y, Jansen W, Greuter M, Nelissen RL, Meeuwisse CM, Nieuwenhuis EE, Mebius RE, Kraal G (2007) Secretory leukoprotease inhibitor in mucosal lymph node dendritic cells regulates the threshold for mucosal tolerance. J Immunol 179(10):6588–6595PubMed
23.
Zurück zum Zitat van Wetering S, van der Linden AC, van Sterkenburg MA, de Boer WI, Kuijpers AL, Schalkwijk J, Hiemstra PS (2000) Regulation of SLPI and elafin release from bronchial epithelial cells by neutrophil defensins. Am J Physiol Lung Cell Mol Physiol 278(1):L51–L58PubMed van Wetering S, van der Linden AC, van Sterkenburg MA, de Boer WI, Kuijpers AL, Schalkwijk J, Hiemstra PS (2000) Regulation of SLPI and elafin release from bronchial epithelial cells by neutrophil defensins. Am J Physiol Lung Cell Mol Physiol 278(1):L51–L58PubMed
24.
Zurück zum Zitat Grobmyer SR, Barie PS, Nathan CF, Fuortes M, Lin E, Lowry SF, Wright CD, Weyant MJ, Hydo L, Reeves F, Shiloh MU, Ding A (2000) Secretory leukocyte protease inhibitor, an inhibitor of neutrophil activation, is elevated in serum in human sepsis and experimental endotoxemia. Crit Care Med 28(5):1276–1282PubMed Grobmyer SR, Barie PS, Nathan CF, Fuortes M, Lin E, Lowry SF, Wright CD, Weyant MJ, Hydo L, Reeves F, Shiloh MU, Ding A (2000) Secretory leukocyte protease inhibitor, an inhibitor of neutrophil activation, is elevated in serum in human sepsis and experimental endotoxemia. Crit Care Med 28(5):1276–1282PubMed
25.
Zurück zum Zitat Jendeberg AL, Stralin K, Hultgren O (2013) Antimicrobial peptide plasma concentrations in patients with community-acquired pneumonia. Scand J Infect Dis 45(6):432–437PubMed Jendeberg AL, Stralin K, Hultgren O (2013) Antimicrobial peptide plasma concentrations in patients with community-acquired pneumonia. Scand J Infect Dis 45(6):432–437PubMed
26.
Zurück zum Zitat Cajander S, Rasmussen G, Tina E, Magnuson A, Soderquist B, Kallman J, Stralin K (2018) Dynamics of monocytic HLA-DR expression differs between bacterial etiologies during the course of bloodstream infection. PLoS One 13(2):e0192883PubMedPubMedCentral Cajander S, Rasmussen G, Tina E, Magnuson A, Soderquist B, Kallman J, Stralin K (2018) Dynamics of monocytic HLA-DR expression differs between bacterial etiologies during the course of bloodstream infection. PLoS One 13(2):e0192883PubMedPubMedCentral
27.
Zurück zum Zitat Rubin DB (1976) Inference and missing data. Biometrika 63(3):581–592 Rubin DB (1976) Inference and missing data. Biometrika 63(3):581–592
28.
Zurück zum Zitat Cain DJ, Del Arroyo AG, Ackland GL (2014) Uncontrolled sepsis: a systematic review of translational immunology studies in intensive care medicine. Intensive Care Med Exp 2(1):6PubMedPubMedCentral Cain DJ, Del Arroyo AG, Ackland GL (2014) Uncontrolled sepsis: a systematic review of translational immunology studies in intensive care medicine. Intensive Care Med Exp 2(1):6PubMedPubMedCentral
29.
Zurück zum Zitat Abe R, Oda S, Sadahiro T, Nakamura M, Hirayama Y, Tateishi Y, Shinozaki K, Hirasawa H (2010) Gram-negative bacteremia induces greater magnitude of inflammatory response than Gram-positive bacteremia. Crit Care 14(2):R27PubMedPubMedCentral Abe R, Oda S, Sadahiro T, Nakamura M, Hirayama Y, Tateishi Y, Shinozaki K, Hirasawa H (2010) Gram-negative bacteremia induces greater magnitude of inflammatory response than Gram-positive bacteremia. Crit Care 14(2):R27PubMedPubMedCentral
30.
Zurück zum Zitat Surbatovic M, Popovic N, Vojvodic D, Milosevic I, Acimovic G, Stojicic M, Veljovic M, Jevdjic J, Djordjevic D, Radakovic S (2015) Cytokine profile in severe Gram-positive and Gram-negative abdominal sepsis. Sci Rep 5:11355PubMedPubMedCentral Surbatovic M, Popovic N, Vojvodic D, Milosevic I, Acimovic G, Stojicic M, Veljovic M, Jevdjic J, Djordjevic D, Radakovic S (2015) Cytokine profile in severe Gram-positive and Gram-negative abdominal sepsis. Sci Rep 5:11355PubMedPubMedCentral
31.
Zurück zum Zitat Hahn WO, Mikacenic C, Price BL, Harju-Baker S, Katz R, Himmelfarb J, Wurfel MM, Liles WC (2016) Host derived biomarkers of inflammation, apoptosis, and endothelial activation are associated with clinical outcomes in patients with bacteremia and sepsis regardless of microbial etiology. Virulence 7(4):387–394PubMedPubMedCentral Hahn WO, Mikacenic C, Price BL, Harju-Baker S, Katz R, Himmelfarb J, Wurfel MM, Liles WC (2016) Host derived biomarkers of inflammation, apoptosis, and endothelial activation are associated with clinical outcomes in patients with bacteremia and sepsis regardless of microbial etiology. Virulence 7(4):387–394PubMedPubMedCentral
32.
Zurück zum Zitat Nakamura A, Mori Y, Hagiwara K, Suzuki T, Sakakibara T, Kikuchi T, Igarashi T, Ebina M, Abe T, Miyazaki J, Takai T, Nukiwa T (2003) Increased susceptibility to LPS-induced endotoxin shock in secretory leukoprotease inhibitor (SLPI)-deficient mice. J Exp Med 197(5):669–674PubMedPubMedCentral Nakamura A, Mori Y, Hagiwara K, Suzuki T, Sakakibara T, Kikuchi T, Igarashi T, Ebina M, Abe T, Miyazaki J, Takai T, Nukiwa T (2003) Increased susceptibility to LPS-induced endotoxin shock in secretory leukoprotease inhibitor (SLPI)-deficient mice. J Exp Med 197(5):669–674PubMedPubMedCentral
33.
Zurück zum Zitat Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR (2001) Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 29(7):1303–1310PubMed Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR (2001) Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 29(7):1303–1310PubMed
34.
Zurück zum Zitat Duits LA, Tjabringa GS, Aarts NJ, van't Wout JW, Hiemstra PS, Nibbering PH, van Dissel JT (2003) Plasma secretory leukocyte protease inhibitor in febrile patients. Clin Microbiol Infect 9(7):605–613PubMed Duits LA, Tjabringa GS, Aarts NJ, van't Wout JW, Hiemstra PS, Nibbering PH, van Dissel JT (2003) Plasma secretory leukocyte protease inhibitor in febrile patients. Clin Microbiol Infect 9(7):605–613PubMed
35.
Zurück zum Zitat Mihaila A, Tremblay GM (2001) Human alveolar macrophages express elafin and secretory leukocyte protease inhibitor. Z Naturforsch C 56(3–4):291–297PubMed Mihaila A, Tremblay GM (2001) Human alveolar macrophages express elafin and secretory leukocyte protease inhibitor. Z Naturforsch C 56(3–4):291–297PubMed
36.
Zurück zum Zitat Persson LJ, Aanerud M, Hardie JA, Miodini Nilsen R, Bakke PS, Eagan TM, Hiemstra PS (2017) Antimicrobial peptide levels are linked to airway inflammation, bacterial colonisation and exacerbations in chronic obstructive pulmonary disease. Eur Respir J 49(3):1601328 Persson LJ, Aanerud M, Hardie JA, Miodini Nilsen R, Bakke PS, Eagan TM, Hiemstra PS (2017) Antimicrobial peptide levels are linked to airway inflammation, bacterial colonisation and exacerbations in chronic obstructive pulmonary disease. Eur Respir J 49(3):1601328
37.
Zurück zum Zitat Jacobsen LC, Sorensen OE, Cowland JB, Borregaard N, Theilgaard-Monch K (2008) The secretory leukocyte protease inhibitor (SLPI) and the secondary granule protein lactoferrin are synthesized in myelocytes, colocalize in subcellular fractions of neutrophils, and are coreleased by activated neutrophils. J Leukoc Biol 83(5):1155–1164PubMed Jacobsen LC, Sorensen OE, Cowland JB, Borregaard N, Theilgaard-Monch K (2008) The secretory leukocyte protease inhibitor (SLPI) and the secondary granule protein lactoferrin are synthesized in myelocytes, colocalize in subcellular fractions of neutrophils, and are coreleased by activated neutrophils. J Leukoc Biol 83(5):1155–1164PubMed
38.
Zurück zum Zitat Antoniades CG, Khamri W, Abeles RD, Taams LS, Triantafyllou E, Possamai LA, Bernsmeier C, Mitry RR, O'Brien A, Gilroy D, Goldin R, Heneghan M, Heaton N, Jassem W, Bernal W, Vergani D, Ma Y, Quaglia A, Wendon J, Thursz M (2014) Secretory leukocyte protease inhibitor: a pivotal mediator of anti-inflammatory responses in acetaminophen-induced acute liver failure. Hepatology 59(4):1564–1576PubMed Antoniades CG, Khamri W, Abeles RD, Taams LS, Triantafyllou E, Possamai LA, Bernsmeier C, Mitry RR, O'Brien A, Gilroy D, Goldin R, Heneghan M, Heaton N, Jassem W, Bernal W, Vergani D, Ma Y, Quaglia A, Wendon J, Thursz M (2014) Secretory leukocyte protease inhibitor: a pivotal mediator of anti-inflammatory responses in acetaminophen-induced acute liver failure. Hepatology 59(4):1564–1576PubMed
39.
Zurück zum Zitat Markle JG, Fish EN (2014) SeXX matters in immunity. Trends Immunol 35(3):97–104PubMed Markle JG, Fish EN (2014) SeXX matters in immunity. Trends Immunol 35(3):97–104PubMed
40.
Zurück zum Zitat Muenchhoff M, Goulder PJ (2014) Sex differences in pediatric infectious diseases. J Infect Dis 209(Suppl 3):S120–S126PubMedPubMedCentral Muenchhoff M, Goulder PJ (2014) Sex differences in pediatric infectious diseases. J Infect Dis 209(Suppl 3):S120–S126PubMedPubMedCentral
41.
Zurück zum Zitat Bindl L, Buderus S, Dahlem P, Demirakca S, Goldner M, Huth R, Kohl M, Krause M, Kuhl P, Lasch P, Lewandowski K, Merz U, Moeller J, Mohamad Y, Peters M, Porz W, Vierzig A, Ruchard J, Scharf J, Varnholt V (2003) Gender-based differences in children with sepsis and ARDS: the ESPNIC ARDS Database Group. Intensive Care Med 29(10):1770–1773PubMed Bindl L, Buderus S, Dahlem P, Demirakca S, Goldner M, Huth R, Kohl M, Krause M, Kuhl P, Lasch P, Lewandowski K, Merz U, Moeller J, Mohamad Y, Peters M, Porz W, Vierzig A, Ruchard J, Scharf J, Varnholt V (2003) Gender-based differences in children with sepsis and ARDS: the ESPNIC ARDS Database Group. Intensive Care Med 29(10):1770–1773PubMed
42.
Zurück zum Zitat Falagas ME, Mourtzoukou EG, Vardakas KZ (2007) Sex differences in the incidence and severity of respiratory tract infections. Respir Med 101(9):1845–1863PubMed Falagas ME, Mourtzoukou EG, Vardakas KZ (2007) Sex differences in the incidence and severity of respiratory tract infections. Respir Med 101(9):1845–1863PubMed
43.
Zurück zum Zitat Draijer C, Hylkema MN, Boorsma CE, Klok PA, Robbe P, Timens W, Postma DS, Greene CM, Melgert BN (2016) Sexual maturation protects against development of lung inflammation through estrogen. Am J Physiol Lung Cell Mol Physiol 310(2):L166–L174PubMed Draijer C, Hylkema MN, Boorsma CE, Klok PA, Robbe P, Timens W, Postma DS, Greene CM, Melgert BN (2016) Sexual maturation protects against development of lung inflammation through estrogen. Am J Physiol Lung Cell Mol Physiol 310(2):L166–L174PubMed
44.
Zurück zum Zitat King AE, Morgan K, Sallenave JM, Kelly RW (2003) Differential regulation of secretory leukocyte protease inhibitor and elafin by progesterone. Biochem Biophys Res Commun 310(2):594–599PubMed King AE, Morgan K, Sallenave JM, Kelly RW (2003) Differential regulation of secretory leukocyte protease inhibitor and elafin by progesterone. Biochem Biophys Res Commun 310(2):594–599PubMed
45.
Zurück zum Zitat Chotirmall SH, Greene CM, Oglesby IK, Thomas W, O'Neill SJ, Harvey BJ, McElvaney NG (2010) 17Beta-estradiol inhibits IL-8 in cystic fibrosis by up-regulating secretory leucoprotease inhibitor. Am J Respir Crit Care Med 182(1):62–72PubMed Chotirmall SH, Greene CM, Oglesby IK, Thomas W, O'Neill SJ, Harvey BJ, McElvaney NG (2010) 17Beta-estradiol inhibits IL-8 in cystic fibrosis by up-regulating secretory leucoprotease inhibitor. Am J Respir Crit Care Med 182(1):62–72PubMed
46.
Zurück zum Zitat Zheng D, Gui B, Gray KP, Tinay I, Rafiei S, Huang Q, Sweeney CJ, Kibel AS, Jia L (2016) Secretory leukocyte protease inhibitor is a survival and proliferation factor for castration-resistant prostate cancer. Oncogene 35(36):4807–4815PubMed Zheng D, Gui B, Gray KP, Tinay I, Rafiei S, Huang Q, Sweeney CJ, Kibel AS, Jia L (2016) Secretory leukocyte protease inhibitor is a survival and proliferation factor for castration-resistant prostate cancer. Oncogene 35(36):4807–4815PubMed
47.
Zurück zum Zitat Xu W, He B, Chiu A, Chadburn A, Shan M, Buldys M, Ding A, Knowles DM, Santini PA, Cerutti A (2007) Epithelial cells trigger frontline immunoglobulin class switching through a pathway regulated by the inhibitor SLPI. Nat Immunol 8(3):294–303PubMed Xu W, He B, Chiu A, Chadburn A, Shan M, Buldys M, Ding A, Knowles DM, Santini PA, Cerutti A (2007) Epithelial cells trigger frontline immunoglobulin class switching through a pathway regulated by the inhibitor SLPI. Nat Immunol 8(3):294–303PubMed
48.
Zurück zum Zitat Jaureguy F, Carbonnelle E, Bonacorsi S, Clec'h C, Casassus P, Bingen E, Picard B, Nassif X, Lortholary O (2007) Host and bacterial determinants of initial severity and outcome of Escherichia coli sepsis. Clin Microbiol Infect 13(9):854–862PubMed Jaureguy F, Carbonnelle E, Bonacorsi S, Clec'h C, Casassus P, Bingen E, Picard B, Nassif X, Lortholary O (2007) Host and bacterial determinants of initial severity and outcome of Escherichia coli sepsis. Clin Microbiol Infect 13(9):854–862PubMed
Metadaten
Titel
Plasma concentrations of secretory leukocyte protease inhibitor (SLPI) differ depending on etiology and severity in community-onset bloodstream infection
verfasst von
Anna Lange
Sara Cajander
Anders Magnuson
Jonas Sundén-Cullberg
Kristoffer Strålin
Olof Hultgren
Publikationsdatum
14.05.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Clinical Microbiology & Infectious Diseases / Ausgabe 8/2019
Print ISSN: 0934-9723
Elektronische ISSN: 1435-4373
DOI
https://doi.org/10.1007/s10096-019-03567-2

Weitere Artikel der Ausgabe 8/2019

European Journal of Clinical Microbiology & Infectious Diseases 8/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.