• No results found

RAGE and the innate immune response in infection and inflammation - Chapter 8: Systemic and local high mobility group box 1 concentrations during severe infection

N/A
N/A
Protected

Academic year: 2021

Share "RAGE and the innate immune response in infection and inflammation - Chapter 8: Systemic and local high mobility group box 1 concentrations during severe infection"

Copied!
18
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

RAGE and the innate immune response in infection and inflammation

van Zoelen, M.A.D.

Publication date 2009

Link to publication

Citation for published version (APA):

van Zoelen, M. A. D. (2009). RAGE and the innate immune response in infection and inflammation.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

8

Systemic and local HMGB1

concentrations during severe

infection

Critical Care Medicine, 2007

Marieke A.D. van Zoelen1,2, Pierre-François Laterre3, Suzanne Q. van Veen4,

Jan W. van Till4, Xavier Wittebole3, Paul Bresser5, Michael W. Tanck6,

Thierry Dugernier7, Akitoshi Ishizaka8, Marja A. Boermeester4 and Tom van der Poll1,2

1Center for Infection and Immunity Amsterdam (CINIMA), 2Center for Experimental and Molecular Medicine and

Departments of 4Surgery, 5Pulmonology and 6Clinical Epidemiology, Biostatistics and Bioinformatics; Academic

Medical Center, University of Amsterdam, Amsterdam, The Netherlands

3 Department of Critical Care Medicine, St. Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium 7Department of Critical Care Medicine, Cliniques St. Pierre, Ottignies, Belgium

8Department of Medicine, Keio University, School of Medicine, Tokyo, Japan

C

ha

p

te

(3)

Abstract

Objective

High mobility group box 1 (HMGB1) has been implicated as a late mediator

in sepsis. We here sought to determine the extent of HMGB1 release in patients with sepsis stratified to the three most common infectious sources and to determine HMGB1 concentrations at the site of infection during peritonitis or pneumonia.

Design Observational studies in patients and healthy humans challenged with

lipopolysaccharide.

Setting Three intensive care and one clinical research units.

Patients and Subjects

Three patient populations were studied: (i) 51 patients with

sepsis due to pneumonia (29), peritonitis (12) or urinary tract infection (UTI) (10), (ii) 17 patients with peritonitis and (iii) 4 patients with community-acquired pneumonia. In addition, 8 healthy humans were studied after intravenous injection of lipopolysaccharide (4 ng/kg).

Interventions One population of healthy volunteers received lipopolysaccharide

intravenously.

Measurements and Main Results Patients with severe sepsis due to pneumonia

displayed elevated circulating HMGB1 concentrations at both day 0 and 3 after inclusion. Patients with sepsis due to peritonitis had elevated HMGB1 levels at day 0, but not at day 3, whereas UTI was associated with a delayed HMGB1 response with elevated levels only at day 3. HMGB1 concentrations did not differ between survivors and non-survivors and were not correlated to either disease severity or concurrently measured cytokine levels. In line, although intravenous lipopolysaccharide injection clearly elevated plasma cytokine levels, HMGB1 remained undetectable. In patients with peritonitis, HMGB1 concentrations in abdominal fluid were >10-fold higher than in concurrently obtained plasma. In pneumonia patients, HMGB1 levels were higher in bronchoalveolar lavage fluid obtained from the site of infection than in lavage fluid from healthy controls.

Conclusions In severe sepsis the kinetics of HMGB1 release may differ depending on

the primary source of infection. In patients with severe infection, HMGB1 release may predominantly occur at the site of infection.

(4)

Introduction

Sepsis is the second leading cause of death in noncoronary intensive care units and the 10th leading cause of death overall (1, 2). Sepsis afflicts approximately 750,000 patients in the United States each year and - despite modern advances in the care of the critically ill - has been associated with mortality rates of 20%-50%. In sepsis, the lung is the primary source of infection (35-54%), followed by the abdomen (20-28%) and the urinary tract (8-13%) (3-5).

Sepsis is associated with the systemic activation of multiple inflammatory pathways. In earlier years much attention has been focused on the role of proinflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1 in the pathogenesis of sepsis and the organ failure that accompanies this syndrome (6, 7). Characteristically, these cytokine mediators have a short circulating half-life and their release primarily occurs early after the exposure to an infectious challenge. In line, neutralization of endogenous TNF-α or IL-1 only confers protection against lethality during experimental sepsis (or systemic inflammatory response syndrome) when administered prophylactically or very early after the infusion of bacteria (or bacterial products). Several years ago, Wang et al. identified high mobility group box 1 (HMGB1) as a possible late mediator of sepsis (8). Unlike TNF-α and IL-1β, HMGB1 release occurred late after injection of lipopolysaccharide (LPS) into mice, starting only after 8 hours, and elevated circulating HMGB1 remained detectable up to 36 hours thereafter (8). Similarly, experimental abdominal sepsis induced by cecal ligation and puncture was associated with a late (after 18 hours) and sustained (more than 72 hours) release of HMGB1 in the circulation (9). Of importance, death of these animals paralleled the accumulation of HMGB1 in their circulation and delayed administration of neutralizing anti-HMGB1 antibodies conferred protection against lethality caused by high dose LPS administration or cecal ligation and puncture in mice (8-10) and in rats (11). Clinical observational studies have further implicated HMGB1 as a late mediator of sepsis: three investigations have reported elevated serum/plasma HMGB1 concentrations in patients with severe sepsis (8, 12, 13).

Recently, high HMGB1 concentrations were found in the pulmonary epithelial lining fluid of patients with sepsis and acute lung injury, suggesting that this mediator is present at the site of local tissue injury (13). In the present study we sought to determine the extent of systemic HMGB1 release in patients with severe sepsis stratified to the three most common primary sources of infection (lung, abdomen and urinary tract) and to examine local HMGB1 levels, at the site of the infection, in patients with peritonitis and pneumonia.

(5)

Materials and Methods

Patients and design

All studies were approved by the scientific and ethics committees of the Academic Medical Center (Amsterdam, the Netherlands), the St. Luke University Hospital and the St. Pierre’s Hospital (both in Brussels, Belgium). Written informed consent was obtained from all subjects or their relatives. The study included three patient populations: (i) 51 patients with severe sepsis, defined as a known or suspected infection plus a systemic inflammatory response syndrome and failure of at least one organ (14); these patients were admitted to the intensive care unit of either St. Luke University Hospital or St. Pierre’s Hospital at the day the diagnosis of severe sepsis was made and serum was obtained on inclusion (“day 0”) and at 3 days thereafter (“day 3”). Low-dose steroids were given to 35 patients (69%) with refractory septic shock, i.e. only to patients requiring vasopressors

for a prolonged period of a minimum of 6 hours. They were given at a dose of 50 mg every 8 hours, until vasopressors could be weaned but no longer than 7 days and then tapered within 2-3 days. 31 healthy subjects served as controls; (ii) 17 peritonitis patients admitted to the surgical ward of the Academic Medical Center; these patients were part of a previous study examining the impact of peritonitis on coagulation and fibrinolysis (15). They required emergency laparotomy because of peritonitis caused by perforation or infection of a visceral organ, by ischemia/necrosis of part of the gastrointestinal tract or by postoperative peritoneal infection. Abdominal fluid was aspirated with a syringe from an abdominal drain in cavum Douglasi. EDTA anticoagulated blood and abdominal fluid samples were taken at index laparotomy for peritonitis (t = 0) and after 1, 2 and 3 days. Blood and abdominal fluid were centrifuged at 1200 x g and 650 x g, respectively for 10 minutes at 4° C; (iii) four spontaneously breathing patients with unilateral community-aquired pneumonia; these patients were previously described in detail (16). Briefly, they fulfilled the following criteria: fever (temperature > 37.7 °C), a chest radiograph within 2 days after admission suspect for a new unilateral infiltrate, no antibiotic pretreatment and arterial partial pressure of oxygen > 7.5 kilopascals while breathing room air. Ten healthy volunteers not taking any medication served as control subjects. Broncholaveolar lavage was performed in a standardized fashion, according to the guidelines of the American Thoracic Society, by use of a flexible fiberoptic video-bronchoscope as described (16). In addition to these three patient populations, eight healthy males (22.6 ± 0.6 years) were studied after intravenous injection of lipopolysaccharide (Escherichia coli LPS, lot G,

United States Pharmacopeial Convention, Rockville, MD) at a dose of 4 ng/kg. In these subjects EDTA anticoagulated blood was obtained before and 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8 and 24 hours after challenge.

(6)

Measurements and assays

Data were collected prospectively from patient records, patient data management system (at the intensive care unit) and hospital information system. The following variables were collected when appropriate: date of birth, gender, APACHE-II and sepsis-related organ failure assessment (SOFA) scores, presence of septic shock, of organ dysfunction (both defined according to the consensus published in Crit Care Med 1992 (17)) and of coagulopathy, use of steroids, length of intensive care stay and of hospital stay, blood culture results, date of death and etiology of peritonitis. HMGB1 was measured by ELISA with the use of monoclonal antibodies to HMGB1 and with standardization to a curve of recombinant human HMGB1 as described previously (18, 19). Briefly, polystyrene microtiter plates were coated with monoclonal anti-calf HMGB1 antibody. Wells were incubated with bovine serum albumin, washed and the calibrator and samples were added to the wells. After washing, another anti-human HMGB1 peroxidase-conjugated monoclonal antibody (a synthetic peptide was used as immunogen) was added to each well. After another washing step, the luminescence reagent was added to the wells. The luminescence was measured using a microplate luminescence reader. Recovery, calculated from data (n = 10) at several concentrations of purified human HMGB1 added to pooled serum as the ratio of the observed concentration to the expected concentration multiplied by 100%, ranged from 92% to 111% in the HMGB1 ELISA assay (19). The detection limit was 0.3 ng/ml and the coefficient of variation < 3%. Tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, IL-10 and IL-12p70 were determined using a cytometric beads array (CBA) multiplex assay (BD Biosciences, San Jose, CA). Detection limit was 2.5 pg/ml for all 6 cytokines.

Statistical analysis

Data are presented as mean ± SEM, median and 25-75% interquartile ranges (IQRs) or number (percentage) when appropriate. Differences between sepsis groups and between abdominal fluid and plasma were performed with non-parametric repeated measures ANOVA or Kruskall-Wallis test. Differences between time points within groups were compared using the Wilcoxon signed-rank test. HMGB1 levels between survivors and non-survivors were compared with a Mann Whitney U test. Correlations were calculated using Spearman’s rho test. p ≤ .05 was considered statistically significant. Statistical analysis

(7)

Results

Sepsis results in elevated serum HMGB1 levels irrespective of the source

of the infection

The demographic and clinical characteristics of patients with severe sepsis are presented in Table 1. In total 51 patients were included with an overall in-hospital mortality of 45%.

Table 1. Characteristics of patients with severe sepsis, stratified to the source of infection Patients with severe sepsis Patients with sepsis due to Peritonitis Patients with sepsis due to Pneumonia Patients with sepsis due to UTI Characteristic n = 51 n = 12 (24) n = 29 (57) n = 10 (20) Age, years 68 ± 1.6 63 ± 4.0 69 ± 1.9 69 ± 3.6 Male sex 31 (61) 9 (75) 16 (55) 6 (60) APACHE-II score 27 ± 1.1 27 ± 1.9 26 ± 1.5 29 ± 2.6 SOFA-score 10 ± 0.6 10 ± 1.1 9 ± 0.9 11 ± 1.7 Shocka 35 (69) 8 (67) 21 (72) 6 (60) Respiratory failurea 32 (63) 2 (17) 25 (86) 5 (50) Renal failurea 25 (49) 4 (33) 14 (48) 7 (70) Metabolic acidosisa 20 (39) 6 (50) 8 (28) 6 (60) Thrombocytopeniaa 17 (33) 6 (50) 5 (17) 6 (60) INR 1.6 ± 0.1 2.4 ± 0.5 1.3 ± 0.1 1.8 ± 0.3 Use of steroids 25 (56) 8 (67) 13 (45) 4 (40)

ICU stay, days 10 (4-25) 15 (7-29) 10 (4-22) 8 (2-14)

Hospital stay, days 23 (12-41) 30 (13-49) 20 (12-36) 31 (14-52)

Pos. blood culture 23 (45) 6 (50) 11 (38) 6 (60)

Mortality 23 (45) 8 (67) 12 (41) 3 (30)

UTI, urinary tract infection; APACHE, acute physiology and chronic health evaluation; SOFA, sepsis-related organ failure asessment; INR,

International Normalized Ratio; ICU, intensive care unit.

Data are mean ± SEM, median (25%-75% interquartile range) or no. (%) when appropriate.

(8)

The primary source of infection was the lung in 29 patients (52%), the abdomen in 12 patients (24%) and the urinary tract in 10 patients (20%). When compared with healthy controls, sepsis patients demonstrated elevated serum HMGB1 concentrations both on admission (day 0) and at day 3 (Fig. 1A, both p < .005).

Figure 1. Serum HMGB1 levels in patients with severe sepsis. HMGB1 concentrations in serum

from patients with severe sepsis (all patients, panel A, n = 51) and subgroups of sepsis patients with different infectious sources; abdomen (peritonitis, panel B, n = 12), lungs (pneumonia, panel C, n = 29) and urinary tract (UTI, panel D, n = 10) and from healthy controls (n = 31). UTI = urinary tract infection. Data are mean ± SEM. *(**) p < .0(00)5 vs. healthy controls.

(9)

Ta ble 2. C yt ok in e le ve ls of pa tie nt s wi th se ve re se psi s Pa tie nt s wi th se ve re se psi s (a ll) Pa tie nt s wi th se psi s d ue t o P eri to ni ti s Pa tie nt s wi th se psi s d ue t o Pn eu m onia Pa tie nt s wi th se psi s d ue t o U TI n = 51 n = 12 n = 29 n = 10 Da y 0 Da y 3 Da y 0 Da y 3 Da y 0 Da y 3 Da y 0 Da y 3 pg/ ml IL-6 1514 ± 382 *** 1032 ± 274 ***, ## 701 ± 274 *** 1096 ± 733 *** 1423 ± 495 *** 1083 ± 661 ***, # 2050 ± 1255 *** 2075 ± 1982 ** IL -8 1198 ± 367 *** 574 ± 292 ***, ## 447 ± 212 *** 213 ± 96 ** 1111 ± 528 *** 774 ± 526 *, # 1237 ± 849 *** 866 ± 793 * IL -10 145 ± 52 *** 61 ± 27 ***, ## 94 ± 55 *** 29 ± 17 ***, # 132 ± 75 *** 46 ± 29 *** 146 ± 105 *** 196 ± 182 *** A ll c yt ok ines w er e mea su re d i n se ru m a t t he d ay of i ncl usio n ( da y 0) a nd 3 d ays t he re aft er ( da y 3). D at a a re mea n ± S EM. U T I, u rin ar y t ract i nfe ct io n. C yt ok ine le vels i n hea lth y c on tr ols w er e lo w o r b elo w det ect io n l im it. * p < .05, ** p < .01, *** p < .005 v s. hea lth y c on tr ols # p < .05, ## p < .01 v s. d ay 0

(10)

Of the subgroups of sepsis patients with different infectious sources, peritonitis and pneumonia patients displayed elevated serum HMGB1 concentrations at day 0 and day 3, although in the peritonitis subgroup the difference with healthy controls at day 3 did not reach statistical significance (p = .106) possibly related to a large interindividual variation.

Patients with sepsis due to urinary tract infection did not have elevated serum HMGB1 levels at day 0, whereas at day 3 circulating HMGB1 concentrations tended to be higher than in healthy controls (p = .054). There was no apparent correlation between serum

HMGB1 levels and the severity of disease: serum HMGB1 did not correlate with either APACHE II scores (r = -0.254, p = .080) or SOFA scores (r = -0.225, p = .120). In addition,

serum HMGB1 concentrations did not differ between survivors and non-survivors (7.2 ± 3.9 ng/ml and 3.0 ± 1.0 ng/ml, respectively, p = .710). HMGB1 serum levels on day 3 did

not differ between patients who used low-dose steroids and patients who did not (data not shown). To investigate whether serum HMGB1 levels are correlated to circulating cytokine levels in patients with severe sepsis, we measured the serum concentrations of TNF-α, IL-1β, IL-6, IL-8, IL-10 and IL-12p70 at day 0 and 3 (Table 2). TNF-α, IL-1β and IL-12p70 levels were very low or undetectable in patients and not significantly different from healthy controls (data not shown). Serum IL-6, IL-8 and IL-10 concentrations were elevated in sepsis patients at both day 0 and day 3, irrespective of the primary source of infection. However, the levels of none of these cytokines demonstrated a correlation with concurrently measured HMGB1 concentrations (data not shown). To further examine a possible relationship between systemic cytokine release and the appearance of HMGB1 in the circulation, we measured cytokine and HMGB1 levels in eight healthy humans intravenously injected with LPS. Whereas LPS induced profound increases in circulating TNF-α, IL-6, IL-8 and IL-10 concentrations (Fig. 2), HMGB1 remained undetectable up to 24 hours after LPS injection (data not shown). Together, these data suggest that sepsis results in a sustained elevation of serum HMGB1 levels irrespective of the source of the infection and that this response is not related to the severity of disease or systemic activation of the cytokine network.

Peritonitis and pneumonia are associated with local HMGB1 release

Next we wished to investigate whether HMGB1 release occurs at the site of infection. For this we measured HMGB1 in abdominal fluid of patients with peritonitis (n = 17) and in bronchoalveolar lavage fluid of patients with pneumonia (n = 4). The demographic and clinical characteristics of peritonitis patients are presented in Table 3. Patients with pneumonia were described previously (16).None of these patients needed to be intubated and none died. Remarkably, both infections were accompanied by high local HMGB1 concentrations. In patients with peritonitis, HMGB1 concentrations in abdominal fluid were more than 10-fold higher than in concurrently obtained plasma and these local levels remained elevated throughout the 3-day sampling period (Fig. 3A). In patients

(11)

Figure 2. LPS induced cytokine release in healthy humans. Plasma cytokine concentrations in

healthy volunteers (n = 8) intravenously challenged with LPS (4 ng/kg, t = 0 hrs). Data are mean ± SEM of TNF-α (A), IL-6 (B), IL-8 (C) and IL-10 (D).

with pneumonia, HMGB1 concentrations were higher in bronchoalveolar lavage fluid harvested from the infected lung than in lavage fluid obtained from healthy controls (Fig. 3B, p < .005). Local HMGB1 levels did not correlate with local cytokine levels (data not

shown).

Discussion

HMGB1 has been widely studied as a nuclear protein that binds DNA, stabilizes nucleosomes and facilitates gene transcription (20). The re-discovery of HMGB1 as an extracellular protein with cytokine-like activity and its pivotal role as a late mediator of experimental sepsis have prompted clinical studies to characterize the release of HMGB1 in patients with sepsis. We here demonstrate that the levels of HMGB1 are elevated in the circulation of patients with sepsis of various origins. Of these, patients with urinary

(12)

Table 3. Characteristics of patients with peritonitis Patients with Peritonitis Characteristic n = 17 Age, years 61 ± 4.0 Male sex 9 (53) APACHE-II score 14 ± 0.8 SOFA score 7 ± 1.4

ICU stay, days 5 (1-20)

Hospital stay, days 30 (17-53)

28-day Mortality 0 (0) Etiology of peritonitis Perforation 8 (47) Anastomotic leakage 7 (41) Abscess 1 (6) Other 1 (6)

APACHE, acute physiology anc chronic health evaluation; SOFA, sepsis-related organ failure asessment; ICU, intensive care unit. Data are mean ± SEM, median (25%-75% interquartile range) or no. (%).

tract infection seemed to have a delayed appearance of HMGB1 in the circulation when compared with patients with pneumonia or peritonitis. Importantly, patients with peritonitis demonstrated strongly elevated HMGB1 concentrations in abdominal fluid, whereas patients with pneumonia displayed elevated levels in BALF obtained from the infected site. These data show for the first time that in patients with severe infection, HMGB1 release occurs at the primary site of infection.

HMGB1 is a 215 amino acid protein that is highly conserved among species. HMGB1 can be secreted by a variety of cell types including monocytes, macrophages, endothelial cells, enterocytes, pituicytes, dendritic cells and natural killer cells (20). HMGB1 can enhance the production of proinflammatory cytokines by monocytes, activate endothelial cells and cause epithelial-cell barrier leak in the gut (21-23). In animal models of sepsis, the kinetics of HMGB1 release in vivo is delayed and more sustained when compared with

the secretion of proinflammatory cytokines like TNF-α, IL-1β and IL-6 (9, 10). In line, various interventions that inhibit HMGB1 activity or production, such as anti-HMGB1 antibodies - the A-box segment of HMGB1 –, ethyl pyruvate and nicotine reduced sepsis

(13)

Figure 3. Local HMGB1 concentrations during infection. HMGB1 concentrations in abdominal

fluid and plasma from patients with peritonitis (A, n = 17) and in bronchoalveolar lavage fluid from patients with pneumonia (B, n = 4). 10 healthy controls served as controls for the patients with pneumonia. Data are mean ± SEM. ** p < .005 vs. healthy controls, *** p < .001 abdominal fluid vs. plasma.

and/or LPS lethality even if treatment was postponed for many hours up to one day (9-11, 24, 25). Notably, recent investigations have implicated apoptosis of immune cells as a crucial event in the pathogenesis of sepsis, playing a major role in immunosuppression and lethality (26). Interestingly, prevention of lymphocyte apoptosis improved survival after cecal ligation and puncture (27), whereas anti-HMGB1 treatment reduced lethality in the same model without influencing apoptosis (10), indicating that HMGB1 production is downstream of apoptosis in the final common pathway to organ damage in severe sepsis. Together these data strongly suggest that the release of HMGB1 is a relatively late event in sepsis that significantly contributes to an adverse outcome.

(14)

Our findings of elevated serum concentrations of HMBG1 in patients with severe sepsis of various origins confirm and extend previous studies. In their original paper describing HMGB1 as a secreted product mediating LPS lethality, Wang et al. reported elevated serum HMGB1 levels in 25 critically ill patients with severe sepsis; measurements were performed only on admission and patients who died had higher HMGB1 concentrations than those who survived (8). Another more recent study examined the kinetics of HMGB1 release in 64 patients with sepsis (12). In line with our current investigation, circulating HMGB1 concentrations remained elevated in most patients for several days after admission; moreover, consistent with our data no correlation was found between HMGB1 levels and severity of disease (12). A third study reported elevated circulating HMGB1 levels in patients with disseminated intravascular coagulation with varying underlying conditions (28). We here show that patients with sepsis resulting from either pneumonia or peritonitis have elevated serum HMGB1 concentrations on admission and three days later, although in abdominal sepsis patients HMGB1 was not statistically significantly increased anymore, possibly due to a large interindividual variation. In patients with sepsis resulting from the urinary tract, serum HMGB1 was not elevated on admission; in these patients HMGB1 was increased at day 3. Considering that HMGB1 concentrations were similar in survivors and non-survivors and that they were not correlated with APACHE-II or SOFA scores, these differences can not be readily explained by differences in severity of disease. Additional research is warranted to further explore the potential differences in the kinetics of HMGB1 release in patients with sepsis depending on the primary source of the infection. The absence of a correlation between serum HMGB1 and concurrently measured cytokine concentrations suggest that these host responses are not linked. To further investigate this issue we measured the levels of these cytokines and HMGB1 at multiple time points after intravenous injection of LPS into healthy humans. In accordance with the absence of a correlation between circulation levels of cytokines and HMGB1 in sepsis patients, a bolus intravenous injection of LPS did not induce detectable plasma levels of HMGB1 although this challenge elicited profound rises in the levels of TNF-α, IL-6, IL-8 and IL-10.

We here measured immunoreactive HMGB1 which may differ from biologically active HMGB1. It is still unknown whether the currently used HMGB1 assays used by our and other laboratories detect biologically active HMGB1 (29). It has been suggested that the biological activities of HMGB1 could vary depending on its post-translational modifications (30); HMGB1 released passively from necrotic cells is molecularly different from the HMGB1 actively secreted by e.g. inflammatory cells because the latter event

requires acetylation (31). The observed elevated systemic and local HMGB1 levels in patients from our studies could be due to this passive as well as active release of HMGB1. Just like currently used (and published) HMGB1 detection methods by other laboratories, our HMGB1 ELISA does not distinguish between these (and possible other) different

(15)

forms of HMGB1. Further experiments are needed to 1) investigate the biological activity of (different forms of ) HMGB1 and 2) develop HMGB1 ELISA assays which distinguish between these (possibly also functionally) different forms of HMGB1.

To the best of our knowledge local HMGB1 levels at sites of infection have not been reported in patients before. We here report for the first time that both patients with peritonitis and patients with pneumonia displayed elevated levels in fluid obtained from the abdomen and bronchoalveolar space, respectively. In patients with peritonitis, local HMGB1 levels were approximately 10-fold higher than in concurrently obtained plasma. Local release of HMGB1 in the bronchoalveolar compartment has been implicated in the pathogenesis of acute lung injury, i.e. patients and animals with acute lung injury had

elevated pulmonary HMGB1 concentrations and inhibition of HMGB1 attenuated lung inflammation and damage (13, 32-34). It remains to be established to what extent local HMGB1 release contributes to circulating HMGB1 in sepsis and to what extent it contributes to damage at the site of infection and distant organs.

Conclusions

The role of HMGB1 as a late mediator of sepsis has been established by a number of studies in experimental animals. We here expand previous studies on HMGB1 levels in patients with sepsis (8, 12, 13), showing that HMGB1 release occurs in sepsis irrespective of the primary source of infection, although in urinary tract infection HMGB1 secretion into the circulation may be delayed when compared to pneumonia and peritonitis. In patients were severe infection HMGB1 release may predominantly occur at the site of infection.

Acknowledgements

(16)

References

1. Wheeler AP and Bernard GR: Treating Patients With Severe Sepsis. N Engl J Med 1999;

340:207-214

2. Angus DC, Linde-Zwirble WT, Lidicker J, et al: Epidemiology of Severe Sepsis in the United States: Analysis of Incidence, Outcome, and Associated Costs of Care. Crit Care Med 2001; 29:1303-1310

3. Annane D, Aegerter P, Jars-Guincestre MC, et al: Current Epidemiology of Septic Shock: the CUB-Rea Network. Am J Respir Crit Care Med 2003; 168:165-172

4. Abraham E, Laterre PF, Garg R, et al: Drotrecogin Alfa (Activated) for Adults With Severe Sepsis and a Low Risk of Death. N Engl J Med 2005; 353:1332-1341

5. Warren BL, Eid A, Singer P, et al: Caring for the Critically Ill Patient. High-Dose Antithrombin III in Severe Sepsis: a Randomized Controlled Trial. Jama 2001;

286:1869-1878

6. Cohen J: The Immunopathogenesis of Sepsis. Nature 2002; 420:885-891

7. Hotchkiss RS and Karl IE: The Pathophysiology and Treatment of Sepsis. N Engl J Med

2003; 348:138-150

8. Wang H, Bloom O, Zhang M, et al: HMG-1 As a Late Mediator of Endotoxin Lethality in Mice. Science 1999; 285:248-51

9. Yang H, Ochani M, Li J, et al: Reversing Established Sepsis With Antagonists of Endogenous High-Mobility Group Box 1. Proc Natl Acad Sci U S A 2004; 101:296-301

10. Qin S, Wang H, Yuan R, et al: Role of HMGB1 in Apoptosis-Mediated Sepsis Lethality. J Exp Med 2006; 203:1637-1642

11. Suda K, Kitagawa Y, Ozawa S, et al: Anti-High-Mobility Group Box Chromosomal Protein 1 Antibodies Improve Survival of Rats With Sepsis. World J Surg 2006; 30:1755-1762

12. Sunden-Cullberg J, Norrby-Teglund A, Rouhiainen A, et al: Persistent Elevation of High Mobility Group Box-1 Protein (HMGB1) in Patients With Severe Sepsis and Septic Shock. Crit Care Med 2005; 33:564-573

13. Ueno H, Matsuda T, Hashimoto S, et al: Contributions of High Mobility Group Box Protein in Experimental and Clinical Acute Lung Injury. Am J Respir Crit Care Med 2004;

170:1310-1316

14. Levy MM, Fink MP, Marshall JC, et al: 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Crit Care Med 2003; 31:1250-1256

15. van Till JW, Levi M, Bresser P, et al: Early Procoagulant Shift in the Bronchoalveolar Compartment of Patients With Secondary Peritonitis.

J Infect Dis 2006; 194:1331-1339

16. Rijneveld AW, Weijer S, Bresser P, et al: Local Activation of the Tissue Factor-Factor VIIa Pathway in Patients With Pneumonia and the Effect of Inhibition of This Pathway in Murine Pneumococcal Pneumonia. Crit Care Med 2006; 34:1725-1730

(17)

17 American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med 1992; 20: 864-874

18. Yamada S, Inoue K, Yakabe K, et al: High Mobility Group Protein 1 (HMGB1) Quantified by ELISA With a Monoclonal Antibody That Does Not Cross-React With HMGB2. Clin Chem 2003; 49:1535-1537

19. Yamada S, Yakabe K, Ishii J, et al: New high mobility group box 1 assay system. Clinica Chim Acta 2006; 372: 173-178

20. Lotze MT and Tracey KJ: High-Mobility Group Box 1 Protein (HMGB1): Nuclear Weapon in the Immune Arsenal. Nat Rev Immunol 2005; 5:331-342

21. Andersson U, Wang H, Palmblad K, et al: High Mobility Group 1 Protein (HMG-1) Stimulates Proinflammatory Cytokine Synthesis in Human Monocytes. J Exp Med 2000; 192:565-570

22. Fiuza C, Bustin M, Talwar S, et al: Inflammation-Promoting Activity of HMGB1 on Human Microvascular Endothelial Cells. Blood 2003; 101:2652-2660

23. Sappington PL, Yang R, Yang H, et al: HMGB1 B Box Increases the Permeability of Caco-2 Enterocytic Monolayers and Impairs Intestinal Barrier Function in Mice. Gastroenterology 2002; 123:790-802

24. Ulloa L, Ochani M, Yang H, et al: Ethyl Pyruvate Prevents Lethality in Mice With Established Lethal Sepsis and Systemic Inflammation. Proc Natl Acad Sci U S A 2002; 99:12351-12356

25. Wang H, Liao H, Ochani M, et al: Cholinergic Agonists Inhibit HMGB1 Release and Improve Survival in Experimental Sepsis. Nat Med 2004; 10:1216-1221

26. Hotchkiss RS and Nicholson DW: Apoptosis and Caspases Regulate Death and Inflammation in Sepsis. Nat Rev Immunol 2006; 6:813-822

27. Hotchkiss RS, Chang KC, Swanson PE, et al: Caspase Inhibitors Improve Survival in Sepsis: a Critical Role of the Lymphocyte. Nat Immunol 2000; 1:496-501

28. Hatada T, Wada H, Nobori T, et al: Plasma Concentrations and Importance of High Mobility Group Box Protein in the Prognosis of Organ Failure in Patients With Disseminated Intravascular Coagulation. Thromb Haemost 2005; 94:975-979

29. Gaïni S, Pedersen SS, Koldkjaer OG, et al: High mobility group box-1 in patients with suspected community-acquired infections and sepsis: a prospective study. Crit Care 2007; 11:R32

30. Dumitriu IE, Baruah P, Manfredi AA, et al: HMGB1: guiding immunity from within. Trends Immunol 2005; 26:381-387

31. Bonaldi T, Talamo F, Scaffaldi P, et al: Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J 2003; 22:5551–5560

32. Abraham E, Arcaroli J, Carmody A, et al: HMG-1 As a Mediator of Acute Lung Inflammation. J Immunol 2000; 165:2950-2954

(18)

33. Kim JY, Park JS, Strassheim D, et al: HMGB1 Contributes to the Development of Acute Lung Injury After Hemorrhage. Am J Physiol Lung Cell Mol Physiol 2005; 288:958-965 34. Ogawa EN, Ishizaka A, Tasaka S, et al: Contribution of High Mobility Group Box-1 to

the Development of Ventilator-Induced Lung Injury. Am J Respir Crit Care Med 2006; 17:400-407

Referenties

GERELATEERDE DOCUMENTEN

The findings obtained in this dissertation shed a first light on factors promoting parochial and universal cooperation. This dissertation thus provides useful

Het gebouw, ook wel de Tabakgarage genoemd, verrees hier tussen 1966 en 1971 naar ontwerp van het architectenbureau Zanstra, Gmelig Meyling, De Clercq, Zubli in opdracht

Brief description: Cross-validation of Fauna Europaea (version 2.6.2) and various Orthopteroid species data sets (version 5.0/5.0), including Orthoptera Species File (

69 geopolitical units have been thus considered (see Table 1 ). Other ecological or biogeographical subdivision systems of the European mainland could have been envisaged, but are

Reis AC, Gondim MGC Jr, de Moraes GJ, Hanna R, Schausberger P, Lawson-Balagbo LE, Barros R (2008) Population dynamics of Aceria guerreronis Keifer (Acari: Eriophyidae) and

However, no e¡ect of annexin V binding was observed on the product desorption rates from PS/PE/PC membranes containing 10 mole % of lysoPC and oleic acid (result not shown). It has

And al- though GM volume in both areas correlated (negatively) with severity of dyslexia, only the LOFG correlated with behavioural measures: nega- tively with whole-word reading

Post hoc analysis revealed that the region in blue (LIPL) is smaller in subjects with dyslexia and that the regions in red (LOFG and ROFG) are larger in subjects with