Skip to main content

Main menu

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Podcasts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Podcasts
  • Subscriptions

Human lipopolysaccharide models provide mechanistic and therapeutic insights into systemic and pulmonary inflammation

Daniel Brooks, Laura C. Barr, Sarah Wiscombe, Daniel F. McAuley, A. John Simpson, Anthony J. Rostron
European Respiratory Journal 2020 56: 1901298; DOI: 10.1183/13993003.01298-2019
Daniel Brooks
1Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laura C. Barr
2Dept of Respiratory Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sarah Wiscombe
1Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Daniel F. McAuley
3School of Medicine, Dentistry and Biomedical Sciences, Institute for Health Sciences, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Daniel F. McAuley
A. John Simpson
1Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for A. John Simpson
Anthony J. Rostron
1Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Anthony J. Rostron
  • For correspondence: anthony.rostron@newcastle.ac.uk
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Inflammation is a key feature in the pathogenesis of sepsis and acute respiratory distress syndrome (ARDS). Sepsis and ARDS continue to be associated with high mortality. A key contributory factor is the rudimentary understanding of the early events in pulmonary and systemic inflammation in humans, which are difficult to study in clinical practice, as they precede the patient's presentation to medical services. Lipopolysaccharide (LPS), a constituent of the outer membrane of Gram-negative bacteria, is a trigger of inflammation and the dysregulated host response in sepsis. Human LPS models deliver a small quantity of LPS to healthy volunteers, triggering an inflammatory response and providing a window to study early inflammation in humans. This allows biological/mechanistic insights to be made and new therapeutic strategies to be tested in a controlled, reproducible environment from a defined point in time. We review the use of human LPS models, focussing on the underlying mechanistic insights that have been gained by studying the response to intravenous and pulmonary LPS challenge. We discuss variables that may influence the response to LPS before considering factors that should be considered when designing future human LPS studies.

Abstract

Human lipopolysaccharide challenge models are a useful adjunct to clinical studies in refining understanding of the host innate immune response to help identify therapeutic targets in an attempt to improve outcomes in critical illness https://bit.ly/3aiCmrx

Introduction

Systemic inflammation is central to the pathogenesis of sepsis, which carries a mortality of up to 25% [1]. Acute respiratory distress syndrome (ARDS), which has a mortality of 2–40%, is characterised by acute pulmonary neutrophilic inflammation accompanied by systemic inflammation [2].

Characterising sepsis and ARDS is hampered by incomplete understanding of the pathogenesis of acute systemic and pulmonary inflammatory responses in humans. Clinically, the earliest host responses can rarely be studied, as patients develop illness before presentation. Animal (particularly rodent) models provide some insight but there are differences in the immune responses in human and rodents [3].

Bacterial lipopolysaccharide (LPS, or endotoxin) is a virulence factor of the outer membrane of Gram-negative bacterial cell walls (figure 1) that elicits an inflammatory response [4] (figure 2). Experimental administration of LPS to humans allows access to the initial inflammatory response and how it evolves, thus allowing mechanistic insights into human inflammation to be made. Understanding the mechanism of the response helps to identify potential therapeutic targets.

FIGURE 1
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1

A schematic diagram of the lipopolysaccharide molecule and its location in the cell wall. M: monosaccharide; Ga: galactose; Gl: glucose; H: heptose; KDO: 2-keto-3-deoxyoctonic acid; G: glucosamine; C: carbon.

FIGURE 2
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2

A summarised, simplified version of lipopolysaccharide (LPS) signalling in the human body (for a detailed review see [19, 20, 22]).

In this article, we provide an overview of different human LPS models including the temporal inflammatory changes induced, limitations and challenges, and future considerations for ongoing human LPS studies. Extensive literature around the role of LPS in pulmonary/occupational lung pathology [5] or inhaled LPS models in smokers and atopic patients [6, 7] will not be considered.

Types of human LPS model

LPS models vary in their route of delivery and the inflammatory response generated. A summary of the source and dose of LPS used in the studies referenced in this review is available in tables 1, 2 and 3 for intravenous, pulmonary, and nasal and intradermal administration, respectively.

View this table:
  • View inline
  • View popup
TABLE 1

Summary of source and dose of lipopolysaccharide (LPS) and subject characteristics in studies using intravenous LPS models

View this table:
  • View inline
  • View popup
TABLE 2

Summary of source and dose of lipopolysaccharide (LPS) and subject characteristics in studies using pulmonary LPS models

View this table:
  • View inline
  • View popup
TABLE 3

Summary of source and dose of lipopolysaccharide (LPS) and subject characteristics in studies using nasal and intradermal LPS model

View this table:
  • View inline
  • View popup
TABLE 4

Summary of biochemical/cytokine changes from intravenous lipopolysaccharide (LPS) models

Intravenous LPS triggers production of cytokines and trafficking of leukocytes from and to the systemic circulation [8]; doses of intravenous LPS vary between studies from 0.06 to 4 ng·kg−1. The response is dose dependent [9]: 2 ng and 4 ng·kg−1 doses have been used to induce a more pronounced inflammatory response (2 ng·kg−1 has been validated as eliciting a systemic inflammatory response syndrome [8]), whereas lower doses are used to model conditions in which chronic, low-grade inflammation plays a role, such as type 2 diabetes [10].

Respiratory LPS models exist in two forms: inhaled challenge uses a nebulised saline solution containing LPS, aiming to induce a diffuse pulmonary inflammatory response. Alternatively, LPS can be instilled bronchoscopically, producing a localised response in a bronchopulmonary segment, simulating the inflammatory response in bronchopneumonia. Pathophysiologically, LPS-induced chemokines drive recruitment of activated neutrophils to the lungs, with leak of protein-rich fluid across the alveolar capillary membrane [11], similar to that seen in the hyperinflammatory endotype of ARDS [12].

Dose comparisons between respiratory models are difficult: in contrast with intravenous models, where LPS used is almost uniformly NIH Clinical Centre Reference Endotoxin (CCRE), derived from Escherichia coli 0113, many respiratory studies use 20–60 μg of other commercially available preparations (tables 1, 2 and 3).

Whilst respiratory challenge models provide tissue-specific information, repeated sampling and analysis of the stromal response remains challenging. As such, nasal models, whereby LPS is applied to the mucosa followed by lavage at pre-determined time points, have been developed. These provide an alternative means of analysing local inflammatory responses with less invasive sampling [13]. Doses vary from 10 to 100 μg and tend to use non-CCRE commercial preparations [13–15]. Finally, intra-dermal injection of CCRE-grade LPS has been used as a model to gain insight into the localised inflammatory response in tissue and is the only model that allows detailed insights into the stromal response [16].

LPS structure and chemistry

The LPS molecule is found in the outer membrane of Gram-negative bacteria and is comprised of an O-specific side chain, a core region and lipid A molecule [4, 17, 18] (figure 1). The O-specific side chain is composed of oligosaccharide units attached to the outer region of the carbohydrate core [4, 15]. The inner core contains both heptose and 2-keto-3-deoxyoctonic acid (KDO) and is joined to lipid A by a ketosidic bond [4, 17, 18]. Lipid A is composed of a phosphorylated glucosamine disaccharide backbone with fatty acid chains attached [4, 17, 18]. Lipid A is the immunologically active component of LPS.

LPS stimulates an inflammatory response by binding to cells expressing Toll-like receptor 4 (TLR4). Circulating LPS binds LPS-binding protein (LBP) [19] which is recognised by the membrane-anchored co-receptor, CD14 [19]. This allows LPS to bind to a membrane protein, MD-2, and the extracellular portion of TLR4, which activates two signalling cascades [19, 20]. The Toll-interleukin-1-resistance (TIR) domain recruits adaptor proteins MyD88-adaptor-like protein (Mal) and MyD88, which allows activation of several interleukin (IL)-1 receptor-associated kinases (IRAKs). These IRAKs allow for ubiquitination of the adaptor molecule tumour necrosis factor (TNF)-α receptor-associated factor 6 (TRAF-6) which activates the transforming growth factor β-activated kinase 1 (TAK1) [20–22]. TAK1 phosphorylates several kinases, freeing their respective transcription factors nuclear factor (NF)-κB, cyclic AMP-responsive element-binding protein (CREB) and activator protein 1 (AP1) to translocate into the nucleus [20–22]. In the second pathway, TLR4 is transported to an endosome where it recruits two more adaptor proteins; TIR domain-containing adaptor protein inducing interferon-β (TRIF) and TRIF-related adaptor protein (TRAM) [19, 20, 22]. This complex can recruit TRAF6 and stimulate NF-κB nuclear translocation through TAK1. TRIF ubiquitinates TRAF3, allowing it to recruit two kinases, TANK-binding kinase 1 (TBK1) and IKKi, which phosphorylate interferon regulatory factor 3 (IRF3) [20, 21]. IRF3 translocates to the nucleus and stimulates the production of type I interferons [20–22]. A graphical summary of the LPS signalling pathway is found in figure 2.

Methods

References in this narrative review were selected by a PubMed search using the terms “LPS”, “lipopolysaccharide”, “endotoxin”, “endotoxaemia” and “endotoxemia” from 1990. Results were screened for investigation of biological/clinical endpoints, mechanistic insights or therapeutic agents, and those considered by the authors to be most relevant for building a comprehensive oversight of the effects of LPS in humans were selected for inclusion.

Values for figures 3 and 4 were obtained from either graphs or the body of text from papers identified in the search strategy. These values were used to calculate a mean for each time point and plotted to create a summary display.

FIGURE 3
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3

Graphical summary of the effects of intravenous lipopolysaccharide (LPS) on clinical, haematological, cellular and inflammatory parameters constructed from papers infusing 2 ng·kg−1 Clinical Centre Reference Endotoxin (CCRE) LPS to healthy volunteers. To provide an overview of the effects of intravenous LPS, the average (mean) values of clinically important parameters were calculated from individual papers where data were available for that parameter. Clinical parameters were constructed from yet to be published data from a 2 ng·kg−1 intravenous LPS model within our own lab. For graphs with two x-axes, the parameters displayed on the graph should be read relative to the axis under which they are listed. For example, those listed on the left-hand side of the figure should be read against the left-sided x-axis. Cellular: neutrophils [28, 51, 71, 178]; macrophages [71, 178, 179]; lymphocytes [28, 71, 178, 179]. Cytokine: tumour necrosis factor (TNF)-α [8, 24, 28, 51, 71, 72, 75, 178]; interleukin (IL)-6 [8, 24, 28, 43, 51, 72, 178]; IL-8 [8, 24, 43, 72, 178]; IL-10 [8, 43, 51, 72, 178]; TNF-soluble receptor (SR) [28, 75]. Haematological: F1+2 [40, 43–45, 47, 49]; thrombin–anti-thrombin (TAT) complexes [43–45, 49]; tissue plasminogen activator (tPA) [47, 49, 75]; sTF [39, 47, 73].

FIGURE 4
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4

Graphical summary of the effects of respiratory lipopolysaccharide (LPS) models. Inhaled LPS responses displayed are induced sputum cell counts and induced sputum cytokines. Instilled LPS responses displayed are bronchoalveolar lavage (BAL) cell counts, BAL cytokine levels and BAL haematological parameters. To provide an overview of the effects of respiratory LPS models, the average (mean) values of clinically important parameters were calculated from individual papers where data were available for that parameter. To ensure consistency of responses, only papers using Clinical Centre Reference Endotoxin (CCRE) (20 000 endotoxin units (equivalent to 2 μg) for inhaled papers and 4 ng·kg−1 for instilled papers) were included. Induced sputum cellular: neutrophils [102, 122, 152, 180–182]; macrophages [102, 152, 180, 182]. For graphs with two x-axes, the parameters displayed on the graph should be read relative to the axis under which they are listed. For example, those listed on the left-hand side of the figure should be read against the left-sided x-axis. Induced sputum cytokines: interleukin (IL)-8 [152, 180, 182]; IL-6 [180–182]; tumour necrosis factor (TNF)-α [180, 181]. BAL cellular: neutrophils [109, 111, 113, 114, 118, 183]; macrophages [111, 113, 114, 118]; lymphocytes [113, 114]. BAL cytokines: TNF [111, 113, 114, 118, 183]; IL-6 [111, 113, 114, 118, 183]; IL-8 [111, 113, 114, 118, 183]; TNF-soluble receptor (SR)I [111, 114, 183]. BAL haematological: F1+2 fragments [118]; thrombin–anti-thrombin (TAT) complexes [117–119]; sTF [117–119]; tissue plasminogen activator (tPA) [117, 119].

Intravenous LPS model

Clinical parameters

The clinical response appears within an hour and begins with headache and myalgias, followed by a tachypnoea 2–3 h later [8]. Core temperature rises by 1–3°C with maximal increases seen 3–4 h post-administration [8, 23–27], which coincides with a tachycardia of ∼90–100 beats·min-1 [8, 23, 24, 26–29]. A small decrease in mean arterial pressure is observed by 3 h [26, 29–31]. Temperature and blood pressure normalise by 24 h, whereas heart rate remains mildly elevated [8, 28].

LPS exerts complex cardiovascular effects characterised by a hyperdynamic state but with reduced ventricular contractility. Left ventricle function is impaired relative to saline-loaded controls, and load-independent contractility indices are reduced. This implies that LPS enhances cardiac output through decreased systemic vascular resistance [29–32]. There are reports of transient bradycardic episodes occurring in fasted volunteers following LPS exposure [33, 34]. This is an exaggerated Bezold–Jarisch reflex (a triad of bradycardia, vasodilation and hypotension resulting from stimulation of cardiac receptors as a homeostatic response to hypovolaemia) in a setting of high vagal tone [35]. Many researchers try to prevent this by hydrating volunteers with isotonic crystalloid solutions, this increases intravascular volume but can attenuate the inflammatory response [36].

Haemostatic parameters

Activation of the coagulation system is demonstrated by increased levels of fibrin [37] and tissue factor [38, 39] at 2 h, followed by rises in plasma prothrombin fragments [40–47] and thrombin-antithrombin (TAT) complexes [41–45, 48, 49], which peak at 4 h. This haemostatic response is unaffected by inhibition of IL-6 [45].

Changes in coagulation are followed by upregulation of fibrinolysis, with plasmin–antiplasmin (PAP), tissue plasminogen activator (tPA) and D-dimer levels peaking at 2, 3 and 4 h respectively [41–43, 46, 48, 49]. By 4 h, levels of plasminogen activator inhibitor-1 (PAI-1) rise, indicating a switch to fibrinolytic inhibition [41–43]. Coagulation and fibrinolysis are susceptible to modulation, and both are inhibited by IL-10 [31]. Adrenaline inhibits coagulation but enhances fibrinolysis, demonstrating anti-thrombotic effects in the presence of LPS [50]. All haematological parameters return to baseline by 24 h.

Cellular parameters

Circulating neutrophils initially fall, with a nadir at 1 h, rising to maximal levels around 4–6 h [24, 25, 43, 51]. This can be attenuated by co-administration of IL-1 receptor antagonist [52]. Granulocyte colony-stimulating factor (G-CSF) concentrations peak around 4 h, suggesting neutrophils are mobilised from bone marrow [25, 27, 51]. Absolute monocyte and lymphocyte counts fall, reaching lowest levels between 1 and 3 h [9, 24, 53]; both return to baseline by 24 h.

Molecular changes and LPS tolerance

Epigenetic changes have been described in vitro 1 h after LPS exposure in monocytes; they result in promotion of the pro-inflammatory response and inhibition of differentiation [54, 55]. Histone modifications result in an open chromatin architecture and increased transcription of genes potentiating the inflammatory response [54, 56–58]. Nucleosome remodelling [56], DNA methylation and micro-RNAs also play a role in the transcriptional response to LPS in macrophages in vitro [56, 58].

By 2 h, macrophage expression of transcription factors in vitro has changed significantly [59]. In vivo, the transcriptional response to LPS can be divided into early up-regulated (such as those coding for key inflammatory mediators), late up-regulated (anti-inflammatory products, apoptosis) or down-regulated genes (bio-energetic processes like oxidative phosphorylation) [60–62]. Recently, one group has demonstrated that the transcriptomic response to LPS is dose dependent, identifying 3736 genes differentially regulated by dose. Multivariate modelling showed up-regulated genes again included those coding for TLR4 signalling and interleukin production as well apoptosis, with down-regulated genes coding for lymphocyte signalling pathways. Univariate analysis revealed that nearly 40% of the variance in the dose-dependent gene set was due to changes in leukocyte counts [63]. These findings again highlight that the response to LPS is dose dependent. Furthermore, they show that transcriptomic analysis may be used to unmask subtle variations in LPS response which have previously gone undetected.

Metabolomic analysis of plasma reveals that at 6 h, lipid metabolism is upregulated whilst protein metabolism is suppressed; by 24 h, lipid metabolism is down-regulated and amino acid metabolites are up-regulated [64]. In one study, 15 out of 16 significantly altered metabolites had similar changes in intravenous LPS-challenged individuals and septic patients [65]. Additionally, septic patients who survived shared directionality in 18 out of 20 differentially regulated metabolites at 24 h with LPS-exposed subjects, compared with nine of 20 for non-survivors [65], suggesting that, LPS models mimic the “controlled response leading to recovery” phenotype in sepsis.

Tolerance to LPS, whereby cells exposed to repeated doses display a diminished capacity to generate an inflammatory response [66] is an important feature of intravenous LPS models both in terms of experimental design and in providing insights into immunosuppression in critical illness. Ex vivo and in vivo tolerance have different kinetics: ex vivo tolerance appears rapidly, by 3 h [67], and resolves within 7 days, whereas in vivo tolerance persists for at least 2 weeks [68], abating by 5 weeks [69].

In vitro models demonstrate tolerance is accompanied by enhancement of inhibitory intra-cellular signalling pathways (e.g. p53 signalling), accumulation of transcriptional repressors, the absence of pro-inflammatory activators (e.g. STAT) at gene promoters [54] and down-regulation of TLR4 expression (in vivo) [70].

Biochemical and cytokine parameters

TNF-α [9, 27, 28, 71], interferon (IFN)-γ [72], IL-6 [9, 24, 27, 28, 51] and IL-8 [9, 24, 27, 51] reach peak levels within 1–2 h and return to baseline after 3–6 h (TNF-α and IL-6) [9, 24, 27, 28, 71] or 24 h (IL-8) [9, 27]. Markers of neutrophil degranulation are seen within 6 h [25, 26, 40, 42]. Several chemotactic mediators are also released, macrophage inflammatory protein (MIP-1α) [27], growth-related oncogene (GRO)-α [27, 28], monocyte chemoattractant protein (MCP)-1 [42] and IFN-γ-induced protein (IP)-10 [73] levels peak at 2–5 h. Antecedent oral corticosteroids inhibit the rise in several inflammatory cytokines and chemokines [42], as does blocking P2Y12 receptors [74].

LPS also triggers a compensatory anti-inflammatory response. IL-10 peaks at 3 h [27, 42, 43, 51, 72] with IL-1 receptor antagonist (IL-1Ra) following at 4 h [25, 27, 28, 42, 51]. Induced circulating soluble TNF-α receptor is highest between 2 and 4 h [25, 27, 42, 75]. Several interventions aimed at manipulating the anti-inflammatory response have been trialled using intravenous LPS models. Adrenaline attenuates pro-inflammatory cytokine release [76] whilst enhancing levels of IL-10 [76, 77]. IL-10 release is also increased by the action of nicotine (which stimulates α7-nicotinic acetylcholine receptors, the end point of the vagal anti-inflammatory reflex), highlighting a role for the autonomic nervous system in modulating acute inflammation [78]. Dipyridamole augments anti-inflammatory cytokine secretion [79], which may help explain the association between anti-platelet therapy and improved survival in sepsis [80]. Allogenic adipose mesenchymal stem cell (MSC) infusion, when delivered prior to LPS exposure, has immunomodulatory effects that could be beneficial in sepsis [81].

Following intravenous LPS, the vascular response to noradrenaline [82] is reduced. This is a result of oxidative stress and correctable with high doses of anti-oxidants [82, 83]. Intrinsic catecholamine production is increased however, with adrenaline levels rising by 1–2 h [8, 27, 71]. Endothelial cell activation is triggered by TNF-α [43] and is indicated by maximal blood concentrations of vascular endothelial growth factor (VEGF) [40] and soluble thrombomodulin (sTM) [84] at 3–4 h, von Willebrand factor (vWF) at 5–6 h [26, 48] and E-selectin at 6–12 h [25, 27, 43, 48, 84]. vWF and E-selectin levels are still raised at 24 h [25, 43, 48].

Whilst there is no change in urea and creatinine clearance, renal plasma flow increases 3 h after LPS infusion [85]. Evidence of localised damage is present; levels of cytosolic glutathione-S-transferase-A1 (GSTA1-1), a marker of proximal tubular damage, are raised between 6 and 12 h [86]. Local formation of reactive oxygen and nitrogen species are thought to contribute to this damage, as calcium-insensitive nitric oxide synthase (iNOS) mRNA levels isolated from urinary cells are significantly increased [86]. Novel biomarkers of early kidney injury, such as kidney injury molecule (KIM)1, are also increased by LPS [87]. Treating patients with sepsis/septic shock with alkaline phosphate infusions (which detoxifies lipopolysaccharide) improves renal function by inhibiting upregulation of iNOS and thus the formation of damaging reactive oxygen and nitrogen species [88].

Increases in acute phase proteins are evident at 24 h; levels of LBP have increased 2–5-fold [89–91], serum amyloid-A (SAA) levels by at least 17-fold [92, 93], fibrinogen by approximately 30% [46, 94] and C-reactive protein (CRP) by at least two-fold [9, 24, 72].

Respiratory LPS models

Clinical parameters

Inhaled

Inhalation of LPS raises body temperature by 0.5–1.5°C, peaking 6–8 h post-exposure [95, 96]. Influenza-like symptoms, such as headache and fatigue occur [97, 98]. Changes in blood pressure, respiratory rate or heart rate are uncommon [6, 95]. Significant heterogeneity in forced expiratory volume in 1 s (FEV1) in healthy volunteers following LPS inhalation occurs [99, 100], which may explain why no significant change is demonstrated when evaluating interventions [95, 101–106].

Instilled

Segmental LPS challenge produces minimal clinical symptoms. Some studies have observed a fall in arterial pressure 4 h after exposure [107, 108], decreased arterial oxygen tension and increased arterial carbon dioxide tension [108] or a reduction in FEV1 [107]. Changes in heart rate are variable, with reductions reported at 4 [108] and 24 h [107] but others finding no change [109, 110]. The most consistently reported changes are influenza-like symptoms and increases in temperature of ∼0.5°C by 6 h [108, 109, 111–114].

Haemostatic parameters

Inhaled

At the alveolar level, soluble tissue factor and TAT complexes are found in increased concentrations in bronchoalveolar lavage fluid (BALF) at 6 h [115, 116] whilst protein C activity is reduced [116]. Increased PAI-1 levels are also found, indicating local inhibition of fibrinolysis [115, 116].

Instilled

Instilled LPS results in raised levels of TAT, prothrombin fragments and soluble tissue factor, and depressed protein C levels, in BALF by 6 h [117–119]. Elevated levels of tPA and PAP complexes demonstrate activation of the fibrinolytic system; this is followed by a rise in PAI-1, indicating fibrinolytic depression [117, 119].

Cellular parameters

Inhaled

Evidence of a cellular response is seen in induced sputum samples following LPS inhalation. The sputum neutrophil count peaks between 4 and 6 h and remains elevated at 24 h [95, 102, 120–122], occurring as a result of p38 MAPK-stimulated [123] production of TNF-α, IL-1β and IL-8 [121, 122].

Lymphocyte levels are raised by 24 h and normalise within 7 days [120, 121]. Macrophage levels are variably reported as either static [95, 102, 122] or increased [120, 121]. Biochemical evidence of cell activation mirrors increased cell counts, with the neutrophil enzymes myeloperoxidase (MPO), matrix metalloproteinase (MMP)-9 and human neutrophil elastase (HNE) peaking at 6 h [95, 104, 123, 124].

There is a significant increase in BALF neutrophil count by 90 min [7], which remains elevated at 24 h compared with placebo [106, 125, 126]. Long-acting β-agonists impair alveolar recruitment of neutrophils through inhibition of adenylate cyclase [125]. Alveolar macrophage and lymphocyte counts probably do not increase [106, 125]. There is evidence of increased populations of blood-derived monocytes and dendritic cells subtypes within the alveoli by 8 h; these recruited cells rapidly develop similar gene expression profiles to native airway monocytic cells [127]. Interestingly, LPS is a factor in determining whether a T-helper cell (Th)1 or Th2 response is generated [128], with alveolar macrophages exposed to low doses of inhaled LPS (approximately 2 μg), adopting a Th2 cytokine profile [129]. Inhalation of 60 μg of LPS leads to the appearance of pulmonary monocyte-like cells (PMLCs) in the alveolar spaces [130] with reduced proliferative and phagocytic capacity compared with alveolar macrophages [131].

Systemically, a rise in peripheral circulating neutrophils is seen by 6–8 h [95, 96, 101, 104, 120, 132] which persists to 24 h [96, 101, 120]. The effect on monocytes is unclear: one study found a fall at 6 h [95] and another, using a similar dose, found an increase at 8 h [132]. Lymphocyte numbers are unaffected [95, 132].

Instilled

A large increase in BALF neutrophil count occurs by 6 h, which persists to at least 24 h and is accompanied by smaller increases in macrophage and lymphocyte numbers [108, 113, 133]. Neutrophils recruited to the alveoli have a transcriptomic profile distinct from circulating neutrophils [134]. Instilled LPS results in a peripheral blood neutrophilia with a decrease in the percentage of lymphocytes and monocytes [108, 110].

Biochemical and cytokine parameters

Inhaled

Levels of IL-6, IL-8 and TNF-α, stimulated through p38 MAPK signalling and subsequent IL-1β production [122, 123], peak at 6 h in induced sputum samples [6, 95, 97, 123], as does secretion of the chemotactic marker MIP-1β [123].

In BALF, TNF-α is detectable at significantly elevated concentrations by 90 min and remains raised out to 24 h [7, 105, 125, 126]. Levels of IL-1β [7] and IL-8 are also raised by 90 min, with IL-8 levels still high at 24 h [7, 125, 126]. This is followed by an increase in IL-6 levels at 3–6 h [105, 125]. Markers of increased neutrophil activity such as MPO and HNE peak at 6–9 h [125, 132, 135]. Various chemoattractant molecules, such as MIP-1β and MCP-1 also rise by 6 h [125, 136].

Systemically, IL-6 and IL-8 rise between 6 and 8 h [122, 123, 132], with no change in TNF-α, IL-1β, IL-10 or MCP-1 [122, 123, 132]. Evidence of neutrophil activation is detectable systemically with HNE [132] and MPO [124] raised at 8 and 24 h respectively. As with intravenous models, several strategies aimed at modifying the inflammatory response have been trialled using inhaled LPS. Statins reduce alveolar neutrophil recruitment and TNF-α, MPO and MMP levels [103], although did not improve outcomes in non-selected ARDS patients [137]. This is possibly because inhaled LPS may only model the hyper-inflammatory endotype of ARDS [138]. Recently, infusion of keratinocyte growth factor (KGF) prior to LPS inhalation has been shown to increase type II pneumocyte proliferation, suggesting it may promote lung repair [139].

Inhaled LPS also results in evidence of activation of endothelial cells locally, with raised levels of soluble thrombomodulin in BALF [116], and systemically, where circulating levels of soluble E-selectin rise by 24 h [96, 104, 140].

By 24 h, a rise in acute phase proteins is seen systemically, with LBP increasing 2–5-fold [99, 104], fibrinogen increasing by approximately 30% [118] and CRP levels rising to between two and 10-times baseline values [104, 120]. Oral corticosteroids have been given in inhaled LPS models, reducing the rise in systemic CRP [104] (though with no effect on cytokine secretion [104, 121]).

No change in systemic haematological markers was observed [116].

Instilled

Both pro- and anti-inflammatory activity is detectable in BALF after segmental LPS instillation (table 4). TNF-α, IL-1β, IL-6, IL-8 and G-CSF peak at 6 h [108, 111, 113, 133] and may remain significantly above baseline by 24 h [108, 113]. IL-1Ra levels rise by 6 h [108]; TNF receptor levels are also raised by this point and remain so at 48 h [108]. ENA-78, lactoferrin, MPO and HNE are similarly elevated by 6 h [108, 111]. Local secretion of chemotactic molecules occurs by 6 h [108, 111]. By 12 h, neutrophil-derived MMP-8 and β-defensin-2 concentrations are increased [133]. This pattern of alveolar response parallels the hyper-inflammatory endotype in ARDS [138].

Evidence of endothelial activation following instilled LPS is variable, one study found soluble thrombomodulin in significantly raised quantities in BALF at 6 h [117], whilst, in another, no increase was detected [112].

Systemically, there are significant increases in G-CSF, IL-6 and IL-1Ra over the 24 h following LPS instillation, with CRP levels rising at 24 h [108, 110].

The development of tolerance is less clear in respiratory models, with alveolar macrophages exposed to bronchoscopically instilled LPS in vivo, producing higher levels of inflammatory cytokines upon subsequent ex vivo LPS stimulation than macrophages from the contralateral lung exposed to saline [141].

Challenges and limitations of human LPS models

Most intravenous LPS models involve healthy young males given a single dose of LPS early in the morning. However, the response to intravenous LPS in healthy young males is not the same as other subjects. Females have higher levels of TNF-α [89, 142], CRP [89], cortisol [142] and IL-6 [142], and a greater fall in mean blood pressure [89], whilst also avoiding the loss of sensitivity to noradrenaline that occurs in males [89] in response to LPS. Participation by older subjects is limited, but increasing age is associated with a prolonged fever response and more rapid rise in concentrations of TNF-α [143], and a greater fall in systolic blood pressure [144]. Body mass index (BMI) has not been shown to affect the response to experimental LPS [145, 146], although the range of BMIs assessed has been limited.

Racial differences in the response also exist: Duffy antigen negative Africans have reduced thrombin formation and generate fewer TAT complexes, F1+2 fragments and prothrombin fragments [147], with lower levels of chemokines MCP-1 and GRO-α than Duffy antigen positive Caucasians (though no changes in circulating leukocyte or pro-/anti-inflammatory cytokine levels are found) [148]. In general, people of African ancestry have lower cytokine and CRP responses to LPS than those of European ancestry [92].

Diurnal variation of should also be considered: secretion of several cytokines in response to LPS is higher at night [149].

Additionally, most intravenous models deliver LPS as a single bolus, which may not accurately represent prolonged stimuli in critical illness. Similarly, sepsis and ARDS have a range of causes with variable outcomes whereas LPS is a single insult that represents only one of the virulence factors of Gram-negative bacteria. Additionally, models will not account for the modification of the clinical picture caused by interventions such as antibiotics, fluid resuscitation and organ support [150]. Current models also tend to give interventions prior to LPS exposure, which poorly reflects the temporal relationship of illness and therapy. Finally, the magnitude of the inflammatory response that can be induced in human volunteers is obviously restricted by ethical constraints.

For inhaled LPS models, the use of commercial LPS sources could obscure the dose–response relationship, though some more recent models have used CCRE LPS, generally at a dose of 20 000 endotoxin units (EU) (equivalent to 2 μg) (see table 5). Inhaled LPS models are subject to greater experimental variation than others; different nebulisers produce particles of different sizes [140] and different lung distributions [151] which affects the exposure of lung tissue.

View this table:
  • View inline
  • View popup
TABLE 5

Summary of biochemical/cytokine changes from inhaled/instilled lipopolysaccharide (LPS) models

As such, current experimental design models may not fully encapsulate the heterogeneity of relevant patient populations. Refinement of LPS models should ensure they are not overly reductionist and remain clinically relevant. Furthermore, LPS challenge may only model a specific endotype of a condition (e.g. hyper-inflammatory ARDS) which partially explain the failure to translate therapies demonstrating efficacy in LPS models into successful treatments for critical illness.

Reproducibility of response to LPS

Human LPS models are considered to provide a means of generating a controlled inflammatory response in a reproducible manner. Both bronchoscopic and inhaled LPS respiratory challenges have been shown to produce consistent intra-subject cytokine responses to recurrent challenges (regardless of time between exposures for inhaled LPS) [114, 152, 153]. In intravenous models, similar ratios of induced cytokines have been demonstrated in the same subjects when the exposures are separated by at least 5 weeks [69].

A review of the literature demonstrates that across thousands of volunteers, exposure to LPS generates a predictable response, with localised/systemic neutrophilia and induction of pro- and anti-inflammatory cytokines to broadly similar levels. This is consistent with the personal experience of authors of this review. However, even when models are restricted to phenotypically similar subjects there exists the potential for inter-subject variability. Variable inflammatory responses are an important aspect of human LPS models, as they may help us understand why some patients develop conditions such as ARDS, whilst others, under the same circumstances, do not.

Underlying genetic differences may contribute to this variability, though the relationship is complex; neither the well-characterised TLR4 receptor co-segregating missense mutations Asp299Gly and Thr399Ile nor a number of other frequently studied polymorphisms in genes coding for TNF-α, IFN-γ and IL-10 have effect the response to intravenous LPS [154–156] (though the TLR4 mutations do result in a reduced acute phase protein and leukocyte response after LPS inhalation [157]).

Polymorphisms altering the TNF-α response have perhaps been most focussed on. Mutations in the genes coding for MMP-8 (259A/G), resulting in suppressed MMP-8 production [158], CRP (+1444C/T polymorphism) [159], the β-chain of the fibrinogen molecule (-148C/T polymorphism) [160], result in lower TNF-α levels, whereas mutations in apolipoprotein E alleles cause significantly higher levels of TNF-α to be produced [161].

The cellular and haemostatic responses are also subject to genetic variation: a mutation in the gene coding for E-selectin (561A/C) results in significantly higher F1+2 and D-dimer fragment generation [162], as does the +1444C/T CRP mutation [159]. People who are homozygous for a 163G/T mutation in the coagulation factor XIII-A gene subunit have a less pronounced neutrophil response and smaller fall in circulating monocyte numbers in response to intravenous LPS [163], whilst the E-selectin polymorphism results in monocyte counts 20% higher at 24 h [162].

The response to inhaled LPS also has a genetic component, potentially linked to a deletion polymorphism in an anti-oxidant gene, glutathione-S-transferase Mu1 (GSTM1), present in 40–50% of the population, resulting in increased circulating platelet and total leukocyte counts, and increased sputum neutrophil counts, in response to inhaled LPS [164].

Future considerations

LPS models do not replicate critical illness, although some similar physiological alterations are observed [65] and changes in clinical and inflammatory parameters have been shown to be broadly similar in LPS models and sepsis [23, 165]. When considering lung injury, raised levels of chemotactic mediators (such as MCP-1) and newly characterised inflammatory proteins like whey acidic protein four-disulfide-core 12 (WFDC12) have been demonstrated in healthy volunteers challenged with LPS and ARDS [136, 166].

LPS models provide an important link between basic science and clinical trials, offering the ability to generate an inflammatory response from a defined and accessible time point in human biological systems in a safe, reproducible manner [114, 152, 153].

In vivo LPS models allow inflammation and its interaction with other organ systems, such as cardiovascular or neuro-endocrine responses, to be studied in the whole organism. The findings have greater inference than studies from isolated blood components. For example, it has been shown that ex vivo cytokine production cannot be used to predict the in vivo inflammatory response [167] and that endotoxin tolerance follows different kinetics in in vivo models compared with ex vivo ones [68].

Furthermore, they are useful for proof of concept studies; negative findings in LPS models should give rise to consideration about whether further investigation is warranted.

Future research may benefit from several adjustments to the LPS models currently in use. Standardisation of the source of LPS, dosing criteria and variables measured to assess response, would help comparisons between studies. Expanding models to include study cohorts comprised of women, older subjects and those with chronic diseases may make observations more generalisable.

LPS tolerance must be taken into account when considering experimental design, particularly in intravenous challenge. Appropriate washout periods must be used. Repeated LPS doses are most often given in the investigation of tolerance kinetics [26, 69] or during cross-over studies; generally these include a washout period of >5 weeks [38, 41, 90, 168]. However, several studies have given repeated doses of intravenous LPS with washout periods <2 weeks [83, 85] or not specified [169], suggesting observations could be affected by a tolerance. Induction of LPS tolerance can be used to study the immunosuppressed phenotype of critical illness, with various groups demonstrating it can be reversed with IFN-γ [170] or aspirin [171].

Infusing LPS over several hours may better represent ongoing pathogen exposure. A bolus-followed-by-infusion model resulted in higher cytokine levels and longer duration of pyrexia compared with a bolus of 1 ng·kg−1 (although the total amount of LPS delivered was three-times greater) [172]. Low doses (0.3 ng·kg−1) produce an earlier and more pronounced inflammatory response when delivered via a bolus compared with an infusion [173]. Several newer studies have used an LPS infusion over 4 h [174–177]. Further exploration of these models may allow the evaluation of interventions in the setting of ongoing inflammation and immunosuppression that occurs in sepsis, though repeated exposure places more onerous requirements on both volunteers and researchers. Another strategy may be greater use of models that deliver the intervention after LPS challenge, more accurately replicating real life. This may lead to more rapid exclusion of treatments which only work when applied in an antecedent manner. Finally, rapid advances in “-omics” technology will provide opportunities to dissect the human inflammatory response using LPS, and to compare these responses with endotypes derived from observational cohorts.

Shareable PDF

Supplementary Material

This one-page PDF can be shared freely online.

Shareable PDF ERJ-01298-2019.Shareable

Acknowledgements

We are grateful for the assistance of Aaron Gardner and Laura Geyer, of laurageyerdesign.com, in drafting and designing the figures in this manuscript.

Footnotes

  • Conflict of interest: D. Brooks has nothing to disclose.

  • Conflict of interest: L.C. Barr has nothing to disclose.

  • Conflict of interest: S. Wiscombe has nothing to disclose.

  • Conflict of interest: D.F. McAuley reports personal fees for consultancy from Peptinnovate, Bayer, GlaxoSmithKline, Boehringer Ingelheim and SOBI, grants from NIHR, Wellcome Trust and other funders, institutional payments for undertaking bronchoscopy from GlaxoSmithKline, outside the submitted work; and has a patent for novel treatment for ARDS issued to Queen's University Belfast.

  • Conflict of interest: A.J. Simpson has nothing to disclose.

  • Conflict of interest: A.J. Rostron has nothing to disclose.

  • Received July 1, 2019.
  • Accepted March 18, 2020.
  • Copyright ©ERS 2020
https://www.ersjournals.com/user-licence

References

  1. ↵
    1. Rowan KM,
    2. Angus DC,
    3. Bailey M, et al.
    Early, goal-directed therapy for septic shock - A patient-level meta-analysis. N Engl J Med 2017; 376: 2223–2234. doi:10.1056/NEJMoa1701380
    OpenUrlCrossRefPubMed
  2. ↵
    1. Bellani G,
    2. Laffey JG,
    3. Pham T, et al.
    Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA 2016; 315: 788–800. doi:10.1001/jama.2016.0291
    OpenUrlCrossRefPubMed
  3. ↵
    1. Fink M
    . Animal models of sepsis. Virulence 2014; 5: 143–153. doi:10.4161/viru.26083
    OpenUrlCrossRefPubMed
  4. ↵
    1. Bayston K CJ
    . Bacterial endotoxin and current concepts in the diagnosis and treatment of endotoxaemia. J Med Microbiol 1990; 31: 73–83. doi:10.1099/00222615-31-2-73
    OpenUrlCrossRefPubMedWeb of Science
  5. ↵
    1. Michel O
    . Role of lipopolysaccharide (LPS) in asthma and other pulmonary conditions. J Endotoxin Res 2003; 9: 293–300. doi:10.1177/09680519030090050401
    OpenUrlCrossRefPubMed
  6. ↵
    1. Nightingale JA,
    2. Rogers DF,
    3. Hart LA, et al.
    Effect of inhaled endotoxin on induced sputum in normal, atopic, and atopic asthmatic subjects. Thorax 1998; 53: 563–571. doi:10.1136/thx.53.7.563
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Wesselius LJ,
    2. Nelson ME,
    3. Bailey K, et al.
    Rapid lung cytokine accumulation and neutrophil recruitment after lipopolysaccharide inhalation by cigarette smokers and nonsmokers. J Lab Clin Med 1997; 129: 106–114. doi:10.1016/S0022-2143(97)90167-0
    OpenUrlCrossRefPubMedWeb of Science
  8. ↵
    1. Calvano SE,
    2. Coyle SM
    . Experimental human endotoxemia: a model of the systemic inflammatory response syndrome? Surg Infect (Larchmt) 2012; 13: 293–299. doi:10.1089/sur.2012.155
    OpenUrlCrossRefPubMed
  9. ↵
    1. Suffredini AF,
    2. Hochstein HD,
    3. Mcmahon FG
    . Dose-related inflammatory effects of intravenous endotoxin in humans : evaluation of a new clinical lot of Escherichia coli O: 113 endotoxin. J Infect Dis 1999; 179: 1278–1282. doi:10.1086/314717
    OpenUrlCrossRefPubMedWeb of Science
  10. ↵
    1. Andreasen AS,
    2. Krabbe KS,
    3. Krogh-Madsen R, et al.
    Human endotoxemia as a model of systemic inflammation. Curr Med Chem 2008; 15: 1697–1705. doi:10.2174/092986708784872393
    OpenUrlCrossRefPubMedWeb of Science
  11. ↵
    1. Simpson JL,
    2. Phipps S,
    3. Gibson PG
    . Inflammatory mechanisms and treatment of obstructive airway diseases with neutrophilic bronchitis. Pharmacol Ther 2009; 124: 86–95. doi:10.1016/j.pharmthera.2009.06.004
    OpenUrlCrossRefPubMedWeb of Science
  12. ↵
    1. Proudfoot AG,
    2. McAuley DF,
    3. Griffiths MJD, et al.
    Human models of acute lung injury. Dis Model Mech 2011; 4: 145–153. doi:10.1242/dmm.006213
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Danuser B,
    2. Rebsamen H,
    3. Weber C, et al.
    Lipopolysaccharide-induced nasal cytokine response: A dose-response evaluation. Eur Arch Oto-Rhino-Laryngology 2000; 257: 527–532. doi:10.1007/s004050000285
    OpenUrl
    1. Virtala R,
    2. Ekman AK,
    3. Jansson L, et al.
    Airway inflammation evaluated in a human nasal lipopolysaccharide challenge model by investigating the effect of a CXCR2 inhibitor. Clin Exp Allergy 2012; 42: 590–596. doi:10.1111/j.1365-2222.2011.03921.x
    OpenUrlCrossRefPubMed
  14. ↵
    1. Dhariwal J,
    2. Kitson J,
    3. Jones RE, et al.
    Nasal lipopolysaccharide challenge and cytokine measurement reflects innate mucosal immune responsiveness. PLoS One 2015; 10: e0135363. doi:10.1371/journal.pone.0135363
    OpenUrl
  15. ↵
    1. Basran A,
    2. Jabeen M,
    3. Bingle L, et al.
    Roles of neutrophils in the regulation of the extent of human inflammation through delivery of IL-1 and clearance of chemokines. J Leukoc Biol 2013; 93: 7–19. doi:10.1189/jlb.0512250
    OpenUrlCrossRefPubMed
  16. ↵
    1. Freudenberg MA,
    2. Galanos C
    . Bacterial lipopolysaccharides: Structure, metabolism and mechanisms of action. Int Rev Immunol 1990; 6: 207–221. doi:10.3109/08830189009056632
    OpenUrlCrossRefPubMed
  17. ↵
    1. Rietschel ET,
    2. Kirikae T,
    3. Schade FU, et al.
    The chemical structure of bacterial endotoxin in relation to bioactivity. Immunobiology 1993; 187: 169–190. doi:10.1016/S0171-2985(11)80338-4
    OpenUrlCrossRefPubMedWeb of Science
  18. ↵
    1. Janssens S,
    2. Beyaert R
    . Role of Toll-like receptors in pathogen recognition. Clin Microbiol Rev 2003; 16: 637–646. doi:10.1128/CMR.16.4.637-646.2003
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Kawai T,
    2. Akira S
    . The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 2010; 11: 373–384. doi:10.1038/ni.1863
    OpenUrlCrossRefPubMedWeb of Science
  20. ↵
    1. Lim K-H,
    2. Staudt LM
    . Toll-Like Receptor Signaling Pathways. Cold Spring Harb Perspect Biol 2013; 5: a011247.
    OpenUrlFREE Full Text
  21. ↵
    1. O'Neill LAJ,
    2. Golenbock D,
    3. Bowie AG
    . The history of Toll-like receptors - redefining innate immunity. Nat Rev Immunol 2013; 13: 453–460. doi:10.1038/nri3446
    OpenUrlCrossRefPubMed
  22. ↵
    1. Ostrowski SR,
    2. Berg RMG,
    3. Windeløv NA, et al.
    Coagulopathy, catecholamines, and biomarkers of endothelial damage in experimental human endotoxemia and in patients with severe sepsis: A prospective study. J Crit Care 2013; 28: 586–596. doi:10.1016/j.jcrc.2013.04.010
    OpenUrlCrossRefPubMed
  23. ↵
    1. Dillingh MR,
    2. Van Poelgeest EP,
    3. Malone KE, et al.
    Characterization of inflammation and immune cell modulation induced by low-dose LPS administration to healthy volunteers. J Inflamm 2014; 11: 1–9. doi:10.1186/s12950-014-0028-1
    OpenUrl
  24. ↵
    1. Kuhns DB,
    2. Alvord WG,
    3. Gallin JI
    . Increased circulating cytokines, cytokine antagonists, and e-selectin after intravenous administration of endotoxin in humans. J Infect Dis 1995; 171: 145–152. doi:10.1093/infdis/171.1.145
    OpenUrlCrossRefPubMedWeb of Science
  25. ↵
    1. Draisma A,
    2. Pickkers P,
    3. Bouw MP, et al.
    Development of endotoxin tolerance in humans in vivo. Crit Care Med 2009; 37: 1261–1267. doi:10.1097/CCM.0b013e31819c3c67
    OpenUrlCrossRefPubMedWeb of Science
  26. ↵
    1. Suffredini AF,
    2. Reda D,
    3. Banks SM, et al.
    Effects of recombinant dimeric TNF receptor on human inflammatory responses following intravenous endotoxin administration. J Immunol 1995; 155: 5038–5045.
    OpenUrlAbstract
  27. ↵
    1. Copeland S,
    2. Warren HS,
    3. Lowry SF, et al.
    Acute inflammatory response to endotoxin in mice and humans. Clin Diagn Lab Immunol 2005; 12: 60–67.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Mathru M,
    2. Pollard V,
    3. He G, et al.
    Left ventricular diastolic filling characteristics are not impaired but systolic performance was augmented in the early hours of experimental endotoxemia in humans. Shock 2006; 25: 338–343. doi:10.1097/01.shk.0000209523.05249.27
    OpenUrlCrossRefPubMedWeb of Science
    1. Suffredini AF,
    2. Fromm RE,
    3. Parker MM, et al.
    The cardiovascular response of normal humans to the administration of endtoxin. N Engl J Med 1989; 321: 280–287. doi:10.1056/NEJM198908033210503
    OpenUrlCrossRefPubMedWeb of Science
  29. ↵
    1. Kumar A,
    2. Zanotti S,
    3. Bunnell G, et al.
    Interleukin-10 blunts the human inflammatory response to lipopolysaccharide without affecting the cardiovascular response. Crit Care Med 2005; 33: 331–340. doi:10.1097/01.CCM.0000152229.69180.2
    OpenUrlCrossRefPubMedWeb of Science
  30. ↵
    1. Kumar A,
    2. Bunnell E,
    3. Lynn M, et al.
    Experimental human endotoxemia is associated with depression of load-independent contractility indices: Prevention by the lipid a analogue E5531. Chest 2004; 126: 860–867. doi:10.1378/chest.126.3.860
    OpenUrlCrossRefPubMedWeb of Science
  31. ↵
    1. Williams WV,
    2. Fullerton T,
    3. Fox JC, et al.
    Asystole following endotoxin administration. J Endotoxin Res 2000; 6: 303–306. doi:10.1177/09680519000060040501
    OpenUrlPubMed
  32. ↵
    1. Van Eijk LT,
    2. Pickkers P,
    3. Smits P, et al.
    Severe vagal response after endotoxin administration in humans. Intensive Care Med 2004; 30: 2279–2281. doi:10.1007/s00134-004-2477-0
    OpenUrlCrossRefPubMedWeb of Science
  33. ↵
    1. Campagna JA,
    2. Carter C
    . Clinical relevance of the Bezold-Jarisch reflex. Anesthesiology 2003; 98: 1250–1260. doi:10.1097/00000542-200305000-00030
    OpenUrlCrossRefPubMedWeb of Science
  34. ↵
    1. Dorresteijn MJ,
    2. van Eijk LT,
    3. Netea MG, et al.
    Iso-osmolar prehydration shifts the cytokine response towards a more anti-inflammatory balance in human endotoxemia. J Endotoxin Res 2005; 11: 287–293. doi:10.1177/09680519050110050501
    OpenUrlCrossRefPubMedWeb of Science
  35. ↵
    1. Verbon A,
    2. Joost CMM,
    3. Spek CA, et al.
    Effects of IC14, an anti-CD14 antibody, on coagulation and fibrinolysis during low-grade endotoxemia in humans. J Infect Dis 2003; 187: 55–61. doi:10.1086/346043
    OpenUrlCrossRefPubMed
  36. ↵
    1. Schoergenhofer C,
    2. Schwameis M,
    3. Hobl E-L, et al.
    Potent irreversible P2Y12 inhibition does not reduce LPS-induced coagulation activation in a randomized, double-blind, placebo-controlled trial. Clin Sci 2016; 130: 433–440. doi:10.1042/CS20150591
    OpenUrl
  37. ↵
    1. Lemaire LC,
    2. de Kruif MD,
    3. Giebelen IA, et al.
    Dobutamine does not influence inflammatory pathways during human endotoxemia. Crit Care Med 2006; 34: 1365–1371. doi:10.1097/01.CCM.0000215514.96888.E3
    OpenUrlCrossRefPubMed
  38. ↵
    1. Mittermayer F,
    2. Pleiner J,
    3. Schaller G, et al.
    Marked increase in vascular endothelial growth factor concentrations during Escherichia coli endotoxin-induced acute inflammation in humans. Eur J Clin Invest 2003; 33: 758–761. doi:10.1046/j.1365-2362.2003.01192.x
    OpenUrlCrossRefPubMedWeb of Science
  39. ↵
    1. Pajkrt D,
    2. van der Poll T,
    3. Levi M, et al.
    Interleukin-10 inhibits activation of coagulation and fibrinolysis during human endotoxemia. Blood 1997; 89: 2701–2705. doi:10.1182/blood.V89.8.2701
    OpenUrlAbstract/FREE Full Text
  40. ↵
    1. de Kruif MD,
    2. Lemaire LC,
    3. Giebelen IA, et al.
    Prednisolone dose-dependently influences inflammation and coagulation during human endotoxemia. J Immunol 2007; 178: 1845–1851. doi:10.4049/jimmunol.178.3.1845
    OpenUrlAbstract/FREE Full Text
  41. ↵
    1. van der Poll T C,
    2. Levi M SM,
    3. Jansen PM, et al.
    Effect of a recombinant dimeric tumor necrosis factor receptor on inflammatory responses to intravenous endotoxin in normal humans. Blood 1997; 89: 3727–3734.
    OpenUrlAbstract/FREE Full Text
    1. Leitner JM,
    2. Firbas C,
    3. Mayr FB, et al.
    Recombinant human antithrombin inhibits thrombin formation and interleukin 6 release in human endotoxemia. Clin Pharmacol Ther 2006; 79: 23–34. doi:10.1016/j.clpt.2005.10.003
    OpenUrlCrossRefPubMedWeb of Science
  42. ↵
    1. Derhaschnig U,
    2. Bergmair D,
    3. Marsik C, et al.
    Effect of interleukin-6 blockade on tissue factor-induced coagulation in human endotoxemia. Crit Care Med 2004; 32: 1136–1140. doi:10.1097/01.CCM.0000126265.08175.BE
    OpenUrlCrossRefPubMedWeb of Science
  43. ↵
    1. Pernerstorfer T,
    2. Stohlawetz P,
    3. Hollenstein U, et al.
    Endotoxin-induced activation of the coagulation cascade in humans: Effect of acetylsalicylic acid and acetaminophen. Arterioscler Thromb Vasc Biol 1999; 19: 2517–2523. doi:10.1161/01.ATV.19.10.2517
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Pernerstorfer T,
    2. Hollenstein U,
    3. Hansen JB, et al.
    Heparin blunts endotoxin-induced coagulation activation. Circulation 1999; 100: 2485–2490. doi:10.1161/01.CIR.100.25.2485
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Renckens R,
    2. Weijer S,
    3. De Vos AF, et al.
    Inhibition of plasmin activity by tranexamic acid does not influence inflammatory pathways during human endotoxemia. Arterioscler Thromb Vasc Biol 2004; 24: 483–488. doi:10.1161/01.ATV.0000118280.95422.48
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Pernerstorfer T,
    2. Hollenstein U,
    3. Hansen J-B, et al.
    Lepirudin blunts endotoxin-induced coagulation activation. Blood 2000; 95: 1729–1734. doi:10.1182/blood.V95.5.1729.005k16_1729_1734
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. van der Poll T,
    2. Levi M,
    3. Dentener M, et al.
    Epinephrine exerts anticoagulant effects during human endotoxemia. J Exp Med 1997; 185: 1143–1148. doi:10.1084/jem.185.6.1143
    OpenUrlAbstract/FREE Full Text
  48. ↵
    1. de Kleijn S,
    2. Kox M,
    3. Sama IE, et al.
    Transcriptome kinetics of circulating neutrophils during human experimental endotoxemia. PLoS One 2012; 7: e38255. doi:10.1371/journal.pone.0038255
    OpenUrlCrossRefPubMed
  49. ↵
    1. Granowitz E V,
    2. Porat R,
    3. Mier JW, et al.
    Hematologic and immunomodulatory effects of an interleukin 1 receptor antagonist coinfusion during low dose endotoxemia in healthy humans. Blood 1993; 15: 2985–2990. doi:10.1182/blood.V82.10.2985.bloodjournal82102985
    OpenUrl
  50. ↵
    1. Ronit A,
    2. Plovsing RR,
    3. Gaardbo JC, et al.
    Inflammation-induced changes in circulating t-cell subsets and cytokine production during human endotoxemia. J Intensive Care Med 2017; 32: 77–85. doi:10.1177/0885066615606673
    OpenUrlCrossRefPubMed
  51. ↵
    1. Novakovic B,
    2. Habibi E,
    3. Wang SY, et al.
    β-Glucan reverses the epigenetic state of LPS-induced immunological tolerance. Cell 2016; 167: 1354–1368. doi:10.1016/j.cell.2016.09.034
    OpenUrlCrossRef
  52. ↵
    1. Dominguez-Andres J,
    2. Netea MG
    . Long-term reprogramming of the innate immune system. J Leukoc Biol 2019; 105: 329–338.
    OpenUrl
  53. ↵
    1. Seeley JJ,
    2. Ghosh S
    . Molecular mechanisms of innate memory and tolerance to LPS. J Leukoc Biol 2017; 101: 107–119. doi:10.1189/jlb.3MR0316-118RR
    OpenUrlCrossRefPubMed
    1. van der Heijden CD,
    2. Noz MP,
    3. Joosten LAB, et al.
    Epigenetics and trained immunity. Antioxid Redox Signal 2018; 29: 1023–1040. doi:10.1089/ars.2017.7310
    OpenUrl
  54. ↵
    1. Niller HH,
    2. Minarovits J
    . Patho- epigenetics of infectious disease caused by intracellular bacteria. Adv Exp Med Biol 2016; 879: 107–130. 10.1007/978-3-319-24738-0_6.
    OpenUrlCrossRef
  55. ↵
    1. Roach JC,
    2. Smith KD,
    3. Strobe KL, et al.
    Transcription factor expression in lipopolysaccharide-activated peripheral-blood-derived mononuclear cells. Proc Natl Acad Sci 2007; 104: 16245–16250. doi:10.1073/pnas.0707757104
    OpenUrlAbstract/FREE Full Text
  56. ↵
    1. Nguyen TT,
    2. Foteinou PT,
    3. Calvano SE, et al.
    Computational identification of transcriptional regulators in human endotoxemia. PLoS One 2011; 6: 4–6.
    OpenUrlCrossRef
    1. Kamisoglu K,
    2. Calvano SE,
    3. Coyle SM, et al.
    Integrated transcriptional and metabolic profiling in human endotoxemia. Shock 2014; 42: 499–508. doi:10.1097/SHK.0000000000000248
    OpenUrl
  57. ↵
    1. Talwar S,
    2. Munson PJ,
    3. Barb J, et al.
    Gene expression profiles of peripheral blood leukocytes after endotoxin challenge in humans. Physiol Genomics 2006; 25: 203–215. doi:10.1152/physiolgenomics.00192.2005
    OpenUrlCrossRefPubMedWeb of Science
  58. ↵
    1. Khan HN,
    2. Perlee D,
    3. Schoenmaker L, et al.
    Leukocyte transcriptional signatures dependent on LPS dosage in human endotoxemia. J Leukoc Biol 2019; 106: 1153–1160. doi:10.1002/JLB.4A0219-050R
    OpenUrl
  59. ↵
    1. Kamisoglu K,
    2. Sleight KE,
    3. Calvano SE, et al.
    Temporal metabolic profiling of plasma during endotoxemia in humans. Shock 2013; 40: 519–526. doi:10.1097/SHK.0000000000000063
    OpenUrlCrossRefPubMed
  60. ↵
    1. Kamisoglu K,
    2. Haimovich B,
    3. Calvano SE, et al.
    Human metabolic response to systemic inflammation: assessment of the concordance between experimental endotoxemia and clinical cases of sepsis/SIRS. Crit Care 2015; 19: 71. doi:10.1186/s13054-015-0783-2
    OpenUrlCrossRefPubMed
  61. ↵
    1. Biswas SK,
    2. Lopez-Collazo E
    . Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol 2009; 30: 475–487. doi:10.1016/j.it.2009.07.009
    OpenUrlCrossRefPubMedWeb of Science
  62. ↵
    1. de Vos AF,
    2. Pater JM,
    3. van den Pangaart PS, et al.
    In vivo lipopolysaccharide exposure of human blood leukocytes induces cross-tolerance to multiple TLR ligands. J Immunol 2009; 183: 533–542. doi:10.4049/jimmunol.0802189
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Kox M,
    2. de Kleijn S,
    3. Pompe JC, et al.
    Differential ex vivo and in vivo endotoxin tolerance kinetics following human endotoxemia. Crit Care Med 2011; 39: 1866–1870. doi:10.1097/CCM.0b013e3182190d5d
    OpenUrlCrossRefPubMedWeb of Science
  64. ↵
    1. Rittig N,
    2. Thomsen HH,
    3. Bach E, et al.
    Hormone and cytokine responses to repeated endotoxin exposures no evidence of endotoxin tolerance after 5 weeks in humans. Shock 2015; 44: 32–35. doi:10.1097/SHK.0000000000000384
    OpenUrl
  65. ↵
    1. Marsik C,
    2. Mayr F,
    3. Cardona F, et al.
    Endotoxaemia modulates Toll-like receptors on leucocytes in humans. Br J Haematol 2003; 121: 653–656. doi:10.1046/j.1365-2141.2003.04350.x
    OpenUrlCrossRefPubMedWeb of Science
  66. ↵
    1. Richard RP,
    2. Craig RD,
    3. Fong Y, et al.
    Peripheral blood leukocyte kinetics following in vivo lipopolysaccharide (LPS) administration to normal human subjects. Ann Surg 1989; 210: 239–245. doi:10.1097/00000658-198908000-00018
    OpenUrlCrossRefPubMedWeb of Science
  67. ↵
    1. Kemna E,
    2. Pickkers P,
    3. Nemeth E, et al.
    Time-course analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS. Blood 2005; 106: 1864–1866. doi:10.1182/blood-2005-03-1159
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. de Kruif MD,
    2. Lemaire LC,
    3. Giebelen IA, et al.
    Effects of prednisolone on the systemic release of mediators of cell-mediated cytotoxicity during human endotoxemia. Shock 2008; 29: 458–461. doi:10.1097/SHK.0b013e3181598a6a
    OpenUrlPubMed
  69. ↵
    1. Thomas MR,
    2. Outteridge SN,
    3. Ajjan RA, et al.
    Platelet P2Y12 inhibitors reduce systemic inflammation and its prothrombotic effects in an experimental human model. Arterioscler Thromb Vasc Biol 2015; 35: 2562–2570. doi:10.1161/ATVBAHA.115.306528
    OpenUrlAbstract/FREE Full Text
  70. ↵
    1. van der Poll T,
    2. Coyle S,
    3. Kumar A, et al.
    Down-regulation of surface receptors for TNF and IL-1 on circulating monocytes and granulocytes during human endotoxemia: effect of neutralization of endotoxin-induced TNF activity by infusion of a recombinant dimeric TNF receptor. J Immunol 1997; 158: 1490–1497.
    OpenUrlAbstract
  71. ↵
    1. van der Poll T,
    2. Coyle SM,
    3. Barbosa K, et al.
    Epinephrine inhibits tumor necrosis factor-alpha and potentiates interleukin 10 production during human endotoxemia. J Clin Invest 1996; 97: 713–719. doi:10.1172/JCI118469
    OpenUrlCrossRefPubMedWeb of Science
  72. ↵
    1. Kox M,
    2. van Eijk LT,
    3. Zwaag J, et al.
    Voluntary activation of the sympathetic nervous system and attenuation of the innate immune response in humans. Proc Natl Acad Sci USA 2014; 111: 7379–7384. doi:10.1073/pnas.1322174111
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Wittebole X,
    2. Hahm S,
    3. Coyle SM, et al.
    Nicotine exposure alters in vivo human responses to endotoxin. Clin Exp Immunol 2007; 147: 28–34.
    OpenUrlPubMedWeb of Science
  74. ↵
    1. Ramakers BP,
    2. Riksen NP,
    3. Stal TH, et al.
    Dipyridamole augments the antiinflammatory response during human endotoxemia. Crit Care 2011; 15: R289. doi:10.1186/cc10576
    OpenUrlCrossRefPubMed
  75. ↵
    1. Akinosoglou K,
    2. Alexopoulos D
    . Use of antiplatelet agents in sepsis: A glimpse into the future. Thromb Res 2014; 133: 131–138. doi:10.1016/j.thromres.2013.07.002
    OpenUrlCrossRefPubMed
  76. ↵
    1. Perlee D,
    2. van Vught LA,
    3. Scicluna BP, et al.
    Intravenous infusion of human adipose mesenchymal stem cells modifies the host response to lipopolysaccharide in humans: a randomized, single-blind, parallel group, placebo controlled trial. Stem Cells 2018; 36: 1778–1788. doi:10.1002/stem.2891
    OpenUrl
  77. ↵
    1. Pleiner J,
    2. Mittermayer F,
    3. Schaller G, et al.
    Inflammation-induced vasoconstrictor hyporeactivity is caused by oxidative stress. J Am Coll Cardiol 2003; 42: 1656–1662. doi:10.1016/j.jacc.2003.06.002
    OpenUrlFREE Full Text
  78. ↵
    1. Aschauer S,
    2. Gouya G,
    3. Klickovic U, et al.
    Effect of systemic high dose vitamin C therapy on forearm blood flow reactivity during endotoxemia in healthy human subjects. Vascul Pharmacol 2014; 61: 25–29. doi:10.1016/j.vph.2014.01.007
    OpenUrl
  79. ↵
    1. Jilma B,
    2. Blann A,
    3. Pernerstorfer T, et al.
    Regulation of adhesion molecules during human endotoxemia no acute effects of aspirin. Am J Respir Crit Care Med 1999; 1: 857–863. doi:10.1164/ajrccm.159.3.9805087
    OpenUrl
  80. ↵
    1. Schaller G,
    2. Pleiner J,
    3. Mittermayer F, et al.
    Effects of N-acetylcysteine against systemic and renal hemodynamic effects of endotoxin in healthy humans. Crit Care Med 2007; 35: 1869–1875. doi:10.1097/01.CCM.0000275385.45557.25
    OpenUrlPubMed
  81. ↵
    1. Heemskerk S,
    2. Pickkers P,
    3. Bouw MP, et al.
    Upregulation of renal inducible nitric oxide synthase during human endotoxemia and sepsis is associated with proximal tubule injury. Clin J Am Soc Nephrol 2006; 1: 853–862. doi:10.2215/CJN.00490206
    OpenUrlAbstract/FREE Full Text
  82. ↵
    1. van Poelgeest EP,
    2. Dillingh MR,
    3. de Kam M, et al.
    Characterization of immune cell, endothelial, and renal responses upon experimental human endotoxemia. J Pharmacol Toxicol Methods 2018; 89: 39–46. doi:10.1016/j.vascn.2017.10.004
    OpenUrl
  83. ↵
    1. Heemskerk S,
    2. Masereeuw R,
    3. Moesker O, et al.
    Alkaline phosphatase treatment improves renal function in severe sepsis or septic shock patients. Crit Care Med 2009; 37: 417–423. doi:10.1097/CCM.0b013e31819598af
    OpenUrlCrossRefPubMedWeb of Science
  84. ↵
    1. van Eijk LT,
    2. Dorresteijn MJ,
    3. Smits P, et al.
    Gender differences in the innate immune response and vascular reactivity following the administration of endotoxin to human volunteers. Crit Care Med 2007; 35: 1464–1469. doi:10.1097/01.CCM.0000266534.14262.E8
    OpenUrlCrossRefPubMedWeb of Science
  85. ↵
    1. de Jonge E,
    2. Dekkers PEP,
    3. Creasey AA, et al.
    Tissue factor pathway inhibitor does not influence inflammatory pathways during human endotoxemia. J Infect Dis 2001; 183: 1815–1818. doi:10.1086/320723
    OpenUrlCrossRefPubMedWeb of Science
  86. ↵
    1. Calvano SE,
    2. Thompson WA,
    3. Marra MN, et al.
    Changes in polymorphonuclear leukocyte surface and plasma bactericidal/permeability-increasing protein and plasma lipopolysaccharide binding protein during endotoxemia or sepsis. Arch Surg 1994; 129: 220–226. doi:10.1001/archsurg.1994.01420260116016
    OpenUrlCrossRefPubMedWeb of Science
  87. ↵
    1. Ferguson JF,
    2. Patel PN,
    3. Shah RY, et al.
    Race and gender variation in response to evoked inflammation. J Transl Med 2013; 11: 63. doi:10.1186/1479-5876-11-63
    OpenUrlCrossRefPubMed
  88. ↵
    1. Ferguson JF,
    2. Mulvey CK,
    3. Patel PN, et al.
    Omega-3 PUFA supplementation and the response to evoked endotoxemia in healthy volunteers. Mol Nutr Food Res 2014; 58: 601–613. doi:10.1002/mnfr.201300368
    OpenUrlCrossRefPubMed
  89. ↵
    1. Fong Y,
    2. Marano MA,
    3. Moldawer LL, et al.
    The acute splanchnic and peripheral tissue metabolic response to endotoxin in humans. J Clin Invest 1990; 85: 1896–1904. doi:10.1172/JCI114651
    OpenUrlCrossRefPubMedWeb of Science
  90. ↵
    1. Loh LC,
    2. Vyas B,
    3. Kanabar V, et al.
    Inhaled endotoxin in healthy human subjects: A dose-related study on systemic effects and peripheral CD4+ and CD8+ T cells. Respir Med 2006; 100: 519–528. doi:10.1016/j.rmed.2005.06.003
    OpenUrlPubMed
  91. ↵
    1. Michel O,
    2. Dentener M,
    3. Corazza F, et al.
    Healthy subjects express differences in clinical responses to inhaled lipopolysaccharide that are related with inflammation and with atopy. J Allergy Clin Immunol 2001; 107: 797–804. doi:10.1067/mai.2001.114249
    OpenUrlCrossRefPubMedWeb of Science
  92. ↵
    1. Sundblad BM,
    2. Von Scheele I,
    3. Palmberg L, et al.
    Repeated exposure to organic material alters inflammatory and physiological airway responses. Eur Respir J 2009; 34: 80–88. doi:10.1183/09031936.00105308
    OpenUrlAbstract/FREE Full Text
  93. ↵
    1. Korsgren M,
    2. Linden M,
    3. Entwistle N, et al.
    Inhalation of LPS induces inflammatory airway responses mimicking characteristics of chronic obstructive pulmonary disease. Clin Physiol Funct Imaging 2012; 32: 71–79. doi:10.1111/j.1475-097X.2011.01058.x
    OpenUrlCrossRefPubMed
  94. ↵
    1. Kitz R,
    2. Rose MA,
    3. Borgmann A, et al.
    Systemic and bronchial inflammation following LPS inhalation in asthmatic and healthy subjects. J Endotoxin Res 2006; 12: 367–374. doi:10.1177/09680519060120060701
    OpenUrlCrossRefPubMed
  95. ↵
    1. Kline JN,
    2. Cowden JD,
    3. Hunninghake GW, et al.
    Variable airway responsiveness to inhaled lipopolysaccharide. Am J Respir Crit Care Med 1999; 160: 297–303. doi:10.1164/ajrccm.160.1.9808144
    OpenUrlCrossRefPubMedWeb of Science
  96. ↵
    1. Herbert A,
    2. Carvalheiro M,
    3. Rubenowitz E, et al.
    Reduction of alveolar-capillary diffusion after inhalation of endotoxin in normal subjects. Chest 1992; 102: 1095–1098. doi:10.1378/chest.102.4.1095
    OpenUrlCrossRefPubMedWeb of Science
  97. ↵
    1. Bennett WD,
    2. Alexis NE,
    3. Almond M, et al.
    Effect of inhaled endotoxin on mucociliary clearance and airway inflammation in mild smokers and nonsmokers. J Aerosol Med Pulm Drug Deliv 2014; 27: 459–465. doi:10.1089/jamp.2013.1089
    OpenUrlCrossRefPubMed
  98. ↵
    1. Shyamsundar M,
    2. McKeown STW,
    3. O'Kane CM, et al.
    Simvastatin decreases lipopolysaccharide-induced pulmonary inflammation in healthy volunteers. Am J Respir Crit Care Med 2009; 179: 1107–1114. doi:10.1164/rccm.200810-1584OC
    OpenUrlCrossRefPubMedWeb of Science
  99. ↵
    1. Michel O,
    2. Dentener M,
    3. Cataldo D, et al.
    Evaluation of oral corticosteroids and phosphodiesterase-4 inhibitor on the acute inflammation induced by inhaled lipopolysaccharide in human. Pulm Pharmacol Ther 2007; 20: 676–683. doi:10.1016/j.pupt.2006.08.002
    OpenUrlPubMed
  100. ↵
    1. Roos-Engstrand E,
    2. Wallin A,
    3. Bucht A, et al.
    Increased expression of p38 MAPK in human bronchail epithelium after lipopolysaccharide exposure. Eur Respir J 2005; 25: 797–803. doi:10.1183/09031936.05.00078804
    OpenUrlAbstract/FREE Full Text
  101. ↵
    1. Wallin A,
    2. Pourazar J,
    3. Sandström T
    . LPS-induced bronchoalveolar neutrophilia; effects of salmeterol treatment. Respir Med 2004; 98: 1087–1092. doi:10.1016/j.rmed.2004.03.025
    OpenUrlCrossRefPubMedWeb of Science
  102. ↵
    1. Chen DL,
    2. Rosenbluth DB,
    3. Mintun MA, et al.
    FDG-PET imaging of pulmonary inflammation in healthy volunteers after airway instillation of endotoxin. J Appl Physiol 2006; 100: 1602–1609. doi:10.1152/japplphysiol.01429.2005
    OpenUrlCrossRefPubMedWeb of Science
  103. ↵
    1. O'Grady NP,
    2. Preas HL,
    3. Pugin J, et al.
    Local inflammatory responses following bronchial endotoxin instillation in humans. Am J Respir Crit Care Med 2001; 163: 1591–1598. doi:10.1164/ajrccm.163.7.2009111
    OpenUrlCrossRefPubMedWeb of Science
  104. ↵
    1. Chen DL,
    2. Bedient TJ,
    3. Kozlowski J, et al.
    [18F] Fluorodeoxyglucose positron emission tomography for lung antiinflammatory response evaluation. Am J Respir Crit Care Med 2009; 180: 533–539. doi:10.1164/rccm.200904-0501OC
    OpenUrlCrossRefPubMedWeb of Science
  105. ↵
    1. Plovsing RR,
    2. Berg RMG,
    3. Evans KA, et al.
    Transcompartmental inflammatory responses in humans: IV versus endobronchial administration of endotoxin. Crit Care Med 2014; 42: 1658–1665. doi:10.1097/CCM.0000000000000320
    OpenUrlCrossRefPubMed
  106. ↵
    1. Hoogerwerf JJ,
    2. De Vos AF,
    3. Bresser P, et al.
    Lung inflammation induced by lipoteichoic acid or lipopolysaccharide in humans. Am J Respir Crit Care Med 2008; 178: 34–41. doi:10.1164/rccm.200708-1261OC
    OpenUrlCrossRefPubMedWeb of Science
  107. ↵
    1. Kager LM,
    2. De Boer JD,
    3. Bresser P, et al.
    Intrabronchial activated protein C enhances lipopolysaccharide-induced pulmonary responses. Eur Respir J 2013; 42: 188–197. doi:10.1183/09031936.00057112
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. Hohlfeld JM,
    2. Schoenfeld K,
    3. Lavae-Mokhtari M, et al.
    Roflumilast attenuates pulmonary inflammation upon segmental endotoxin challenge in healthy subjects: A randomized placebo-controlled trial. Pulm Pharmacol Ther 2008; 21: 616–623. doi:10.1016/j.pupt.2008.02.002
    OpenUrlCrossRefPubMed
  109. ↵
    1. Holz O,
    2. Tan L,
    3. Schaumann F, et al.
    Inter- and intrasubject variability of the inflammatory response to segmental endotoxin challenge in healthy volunteers. Pulm Pharmacol Ther 2015; 35: 50–59. doi:10.1016/j.pupt.2015.10.011
    OpenUrl
  110. ↵
    1. Maris NA,
    2. de Vos AF,
    3. Bresser P, et al.
    Activation of coagulation and inhibition of fibrinolysis in the lung after inhalation of lipopolysaccharide by healthy volunteers. Thromb Haemost 2005; 93: 1036–1040. doi:10.1160/TH04-08-0492
    OpenUrlPubMedWeb of Science
  111. ↵
    1. Maris NA,
    2. De Vos AF,
    3. Bresser P, et al.
    Salmeterol enhances pulmonary fibrinolysis in healthy volunteers. Crit Care Med 2007; 35: 57–63. doi:10.1097/01.CCM.0000249827.29387.4E
    OpenUrlCrossRefPubMed
  112. ↵
    1. Hoogerwerf JJ,
    2. de Vos AF,
    3. Levi M, et al.
    Activation of coagulation and inhibition of fibrinolysis in the human lung on bronchial instillation of lipoteichoic acid and lipopolysaccharide. Crit Care Med 2009; 37: 619–625. doi:10.1097/CCM.0b013e31819584f9
    OpenUrlCrossRefPubMedWeb of Science
  113. ↵
    1. Bartko J,
    2. Schoergenhofer C,
    3. Schwameis M, et al.
    Dexamethasone inhibits endotoxin-induced coagulopathy in human lungs. J Thromb Haemost 2016; 14: 2471–2477. doi:10.1111/jth.13504
    OpenUrlCrossRef
  114. ↵
    1. Van Der Poll T,
    2. Levi M,
    3. Nick JA, et al.
    Activated protein C inhibits local coagulation after intrapulmonary delivery of endotoxin in humans. Am J Respir Crit Care Med 2005; 171: 1125–1128. doi:10.1164/rccm.200411-1483OC
    OpenUrlCrossRefPubMedWeb of Science
  115. ↵
    1. Doyen V,
    2. Kassengera Z,
    3. Dinh DHP, et al.
    Time course of endotoxin-induced airways’ inflammation in healthy subjects. Inflammation 2012; 35: 33–38. doi:10.1007/s10753-010-9286-0
    OpenUrl
  116. ↵
    1. Michel O,
    2. Dinh PHD,
    3. Doyen V, et al.
    Anti-TNF inhibits the airways neutrophilic inflammation induced by inhaled endotoxin in human. BMC Pharmacol Toxicol 2014; 15: 60. doi:10.1186/2050-6511-15-60
    OpenUrl
  117. ↵
    1. Hernandez ML,
    2. Mills K,
    3. Almond M, et al.
    IL-1 receptor antagonist reduces endotoxin-induced airway inflammation in healthy volunteers. J Allergy Clin Immunol 2015; 135: 379–385. doi:10.1016/j.jaci.2014.07.039
    OpenUrlCrossRef
  118. ↵
    1. Singh D,
    2. Siew L,
    3. Christensen J, et al.
    Oral and inhaled p38 MAPK inhibitors: effects on inhaled LPS challenge in healthy subjects. Eur J Clin Pharmacol 2015; 71: 1175–1184. doi:10.1007/s00228-015-1920-1
    OpenUrlPubMed
  119. ↵
    1. Thorn J,
    2. Rylander R
    . Inflammatory response after inhalation of bacterial endotoxin assessed by the induced sputum technique. Thorax 1998; 53: 1047–1052. doi:10.1136/thx.53.12.1047
    OpenUrlAbstract/FREE Full Text
  120. ↵
    1. Maris NA,
    2. De Vos AF,
    3. Dessing MC, et al.
    Antiinflammatory effects of salmeterol after inhalation of lipopolysaccharide by healthy volunteers. Am J Respir Crit Care Med 2005; 172: 878–884. doi:10.1164/rccm.200503-451OC
    OpenUrlCrossRefPubMedWeb of Science
  121. ↵
    1. Hecker M,
    2. Linder T,
    3. Ott J, et al.
    Immunomodulation by lipid emulsions in pulmonary inflammation: a randomized controlled trial. Crit Care 2015; 19: 226. doi:10.1186/s13054-015-0933-6
    OpenUrlCrossRefPubMed
  122. ↵
    1. Jardine L,
    2. Wiscombe S,
    3. Reynolds G, et al.
    Lipopolysaccharide inhalation recruits monocytes and dendritic cell subsets to the alveolar airspace. Nat Commun 2019; 10: 1999. doi:10.1038/s41467-019-09913-4
    OpenUrl
  123. ↵
    1. Xu H,
    2. Liew LN,
    3. Kuo IC, et al.
    The modulatory effects of lipopolysaccharide-stimulated B cells on differential T-cell polarization. Immunology 2008; 125: 218–228. doi:10.1111/j.1365-2567.2008.02832.x
    OpenUrlCrossRefPubMed
  124. ↵
    1. Alexis NE,
    2. Lay JC,
    3. Almond M, et al.
    Inhalation of low-dose endotoxin favors local TH2 response and primes airway phagocytes in vivo. J Allergy Clin Immunol 2004; 114: 1325–1331. doi:10.1016/j.jaci.2004.09.002
    OpenUrlCrossRefPubMedWeb of Science
  125. ↵
    1. Brittan M,
    2. Barr L,
    3. Morris AC, et al.
    A novel subpopulation of monocyte-like cells in the human lung after lipopolysaccharide inhalation. Eur Respir J 2012; 40: 206–214. doi:10.1183/09031936.00113811
    OpenUrlAbstract/FREE Full Text
  126. ↵
    1. Brittan M,
    2. Barr LC,
    3. Anderson N, et al.
    Functional characterisation of human pulmonary monocyte-like cells in lipopolysaccharide-mediated acute lung inflammation. J Inflamm (Lond) 2014; 11: 9. doi:10.1186/1476-9255-11-9
    OpenUrl
  127. ↵
    1. Barr LC,
    2. Brittan M,
    3. Morris AC, et al.
    A randomized controlled trial of peripheral blood mononuclear cell depletion in experimental human lung inflammation. Am J Respir Crit Care Med 2013; 188: 449–455. doi:10.1164/rccm.201212-2334OC
    OpenUrlCrossRefPubMedWeb of Science
  128. ↵
    1. Glader P,
    2. Smith ME,
    3. Malmhäll C, et al.
    Interleukin-17-producing T-helper cells and related cytokines in human airways exposed to endotoxin. Eur Respir J 2010; 36: 1155–1164. doi:10.1183/09031936.00170609
    OpenUrlAbstract/FREE Full Text
  129. ↵
    1. Coldren CD,
    2. Nick JA,
    3. Poch KR, et al.
    Functional and genomic changes induced by alveolar transmigration in human neutrophils. Am J Physiol Lung Cell Mol Physiol 2006; 291: L1267–L1276. doi:10.1152/ajplung.00097.2006
    OpenUrlCrossRefPubMedWeb of Science
  130. ↵
    1. Wittkowski H,
    2. Sturrock A,
    3. van Zoelen MA, et al.
    Neutrophil-derived S100A12 in acute lung injury and respiratory distress syndrome. Crit Care Med 2007; 35: 1369–1375. doi:10.1097/01.CCM.0000262386.32287.29
    OpenUrlCrossRefPubMedWeb of Science
  131. ↵
    1. Williams AE,
    2. José RJ,
    3. Mercer PF, et al.
    Evidence for chemokine synergy during neutrophil migration in ARDS. Thorax 2017; 72: 66–73. doi:10.1136/thoraxjnl-2016-208597
    OpenUrlAbstract/FREE Full Text
  132. ↵
    1. McAuley DF,
    2. Laffey JG,
    3. O'Kane CM, et al.
    Simvastatin in the acute respiratory distress syndrome. N Engl J Med 2014; 371: 1695–1703. doi:10.1056/NEJMoa1403285
    OpenUrlCrossRefPubMed
  133. ↵
    1. Calfee CS,
    2. Delucchi K,
    3. Parsons PE, et al.
    Subphenotypes in acute respiratory distress syndrome: Latent class analysis of data from two randomised controlled trials. Lancet Respir Med 2014; 2: 611–620. doi:10.1016/S2213-2600(14)70097-9
    OpenUrl
  134. ↵
    1. Shyamsundar M,
    2. McAuley DF,
    3. Ingram RJ, et al.
    Keratinocyte growth factor promotes epithelial survival and resolution in a human model of lung injury. Am J Respir Crit Care Med 2014; 189: 1520–1529. doi:10.1164/rccm.201310-1892OC
    OpenUrlCrossRefPubMedWeb of Science
  135. ↵
    1. Doyen V,
    2. Pilcer G,
    3. Dinh PHD, et al.
    Inflammation induced by inhaled lipopolysaccharide depends on particle size in healthy volunteers. Br J Clin Pharmacol 2016; 82: 1371–1381. doi:10.1111/bcp.13052
    OpenUrl
  136. ↵
    1. Hoogerwerf JJ,
    2. De Vos AF,
    3. Van't Veer C, et al.
    Priming of alveolar macrophages upon instillation of lipopolysaccharide in the human lung. Am J Respir Cell Mol Biol 2010; 42: 349–356. doi:10.1165/rcmb.2008-0362OC
    OpenUrlCrossRefPubMed
  137. ↵
    1. Engler H,
    2. Benson S,
    3. Wegner A, et al.
    Men and women differ in inflammatory and neuroendocrine responses to endotoxin but not in the severity of sickness symptoms. Brain Behav Immun 2016; 52: 18–26. doi:10.1016/j.bbi.2015.08.013
    OpenUrlCrossRefPubMed
  138. ↵
    1. Krabbe KS,
    2. Bruunsgaard H,
    3. Muff C, et al.
    Ageing Is associated with a prolonged fever response in human endotoxemia ageing is associated with a prolonged fever response in human endotoxemia. Clin Diagn Lab Immunol 2001; 8: 333–338.
    OpenUrlCrossRefPubMed
  139. ↵
    1. Krabbe KS,
    2. Bruunsgaard H,
    3. Qvist J, et al.
    Hypotension during endotoxemia in aged humans. Eur J Anaesthesiol 2001; 18: 572–575. doi:10.1097/00003643-200109000-00003
    OpenUrlCrossRefPubMedWeb of Science
  140. ↵
    1. Rohrer M,
    2. Berger MM,
    3. Pittet YK, et al.
    Metabolic and physiologic effects of an endotoxin challenge in healthy obese subjects. Clin Physiol Funct Imaging 2011; 31: 371–375. doi:10.1111/j.1475-097X.2011.01026.x
    OpenUrlCrossRefPubMed
  141. ↵
    1. van Eijk LT,
    2. van der Pluijm RW,
    3. Ramakers BP, et al.
    Body mass index is not associated with cytokine induction during experimental human endotoxemia. Innate Immun 2014; 20: 61–67. doi:10.1177/1753425913481821
    OpenUrlCrossRefPubMed
  142. ↵
    1. Mayr FB,
    2. Spiel AO,
    3. Leitner JM, et al.
    Racial differences in endotoxin-induced tissue factor-triggered coagulation. J Thromb Haemost 2009; 7: 634–640. doi:10.1111/j.1538-7836.2009.03307.x
    OpenUrlCrossRefPubMedWeb of Science
  143. ↵
    1. Mayr FB,
    2. Spiel AO,
    3. Leitner JM, et al.
    Duffy antigen modifies the chemokine response in human endotoxemia. Crit Care Med 2008; 36: 159–165. doi:10.1097/01.CCM.0000297875.55969.DB
    OpenUrlCrossRefPubMedWeb of Science
  144. ↵
    1. Alamili M,
    2. Bendtzen K,
    3. Lykkesfeldt J, et al.
    Pronounced inflammatory response to endotoxaemia during nighttime: A randomised cross-over trial. PLoS One 2014; 9: e87413. doi:10.1371/journal.pone.0087413
    OpenUrlCrossRefPubMed
  145. ↵
    1. Marshall JC,
    2. Deitch E,
    3. Moldawer LL, et al.
    Preclinical models of shock and sepsis: what can they tell us? Shock 2005; 24: 1–6. doi:10.1097/01.shk.0000191383.34066.4b
    OpenUrlCrossRefPubMed
  146. ↵
    1. Hardy JG,
    2. Newman SP,
    3. Knoch M
    . Lung deposition from four nebulizers. Respir Med 1993; 87: 461–465. doi:10.1016/0954-6111(93)90074-A
    OpenUrlCrossRefPubMedWeb of Science
  147. ↵
    1. Janssen O,
    2. Schaumann F,
    3. Holz O, et al.
    Low-dose endotoxin inhalation in healthy volunteers--a challenge model for early clinical drug development. BMC Pulm Med 2013; 13: 19. doi:10.1186/1471-2466-13-19
    OpenUrl
  148. ↵
    1. Kobernick AK,
    2. Peden DB,
    3. Zhou H, et al.
    Reproducibility of the inflammatory response to inhaled endotoxin in healthy volunteers. J Allergy Clin Immunol 2016; 138: 1205–1207. doi:10.1016/j.jaci.2016.04.017
    OpenUrl
  149. ↵
    1. Kovar FM,
    2. Marsik C,
    3. Cvitko T, et al.
    The tumor necrosis factor α -308 G/A polymorphism does not influence inflammation and coagulation response in human endotoxemia. Shock 2007; 27: 238–241. doi:10.1097/01.shk.0000239768.64786.3a
    OpenUrlCrossRefPubMedWeb of Science
    1. Taudorf S,
    2. Krabbe KS,
    3. Berg RMG, et al.
    Common studied polymorphisms do not affect plasma cytokine levels upon endotoxin exposure in humans. Clin Exp Immunol 2008; 152: 147–152. doi:10.1111/j.1365-2249.2008.03612.x
    OpenUrlPubMed
  150. ↵
    1. Calvano JE,
    2. Bowers DJ,
    3. Coyle SM, et al.
    Response to systemic endotoxemia among humans bearing polymorphisms of the Toll-like receptor 4 (hTLR4). Clin Immunol 2006; 121: 186–190. doi:10.1016/j.clim.2006.05.012
    OpenUrlCrossRefPubMedWeb of Science
  151. ↵
    1. Michel O,
    2. LeVan TD,
    3. Stern D, et al.
    Systemic responsiveness to lipopolysaccharide and polymorphisms in the toll-like receptor 4 gene in human beings. J Allergy Clin Immunol 2003; 112: 923–929. doi:10.1016/j.jaci.2003.05.001
    OpenUrlCrossRefPubMedWeb of Science
  152. ↵
    1. Rella JM,
    2. Jilma B,
    3. Fabry A, et al.
    MMP-8 genotypes influence the inflammatory response in human endotoxemia. Inflammation 2014; 37: 451–456. doi:10.1007/s10753-013-9758-0
    OpenUrlCrossRefPubMed
  153. ↵
    1. Marsik C,
    2. Sunder-Plassmann R,
    3. Jilma B, et al.
    The C-reactive protein +1444C/T alteration modulates the inflammation and coagulation response in human endotoxemia. Clin Chem 2006; 52: 1952–1957. doi:10.1373/clinchem.2006.069823
    OpenUrlAbstract/FREE Full Text
  154. ↵
    1. Kovar FM,
    2. Marsik C,
    3. Jilma B, et al.
    The fibrinogen - 148 C/T polymorphism influences inflammatory response in experimental endotoxemia in vivo. Thromb Res 2007; 120: 727–731. doi:10.1016/j.thromres.2006.12.007
    OpenUrlCrossRefPubMed
  155. ↵
    1. Gale SC,
    2. Gao L,
    3. Mikacenic C, et al.
    APOε4 is associated with enhanced in vivo innate immune responses in human subjects. J Allergy Clin Immunol 2014; 134: 1–8. doi:10.1016/j.jaci.2014.05.028
    OpenUrl
  156. ↵
    1. Jilma B,
    2. Marsik C,
    3. Kovar F, et al.
    The single nucleotide polymorphism Ser128Arg in the E-selectin gene is associated with enhanced coagulation during human endotoxemia. Blood 2005; 105: 2380–2383. doi:10.1182/blood-2004-09-3752
    OpenUrlAbstract/FREE Full Text
  157. ↵
    1. Kovar FM,
    2. Marsik CL,
    3. Jilma B, et al.
    The inflammatory response is influenced by FXIII VAL 34 LEU polymorphism in a human LPS model. Wien Klin Wochenschr 2009; 121: 515–519. doi:10.1007/s00508-009-1182-y
    OpenUrlPubMed
  158. ↵
    1. Dillon MA,
    2. Harris B,
    3. Hernandez ML, et al.
    Enhancement of systemic and sputum granulocyte response to inhaled endotoxin in people with the GSTM1 null genotype. Occup Environ Med 2011; 68: 783–785. doi:10.1136/oem.2010.061747
    OpenUrlAbstract/FREE Full Text
  159. ↵
    1. Cannon JG,
    2. Tompkins RG,
    3. Gelfand JA, et al.
    Circulating interleukin-l and tumor necrosis factor in septic shock and experimental endotoxin fever. J Infect Dis 1990; 161: 79–84. doi:10.1093/infdis/161.1.79
    OpenUrlCrossRefPubMedWeb of Science
  160. ↵
    1. Glasgow AMA,
    2. Small DM,
    3. Scott A, et al.
    A role for whey acidic protein four-disulfide-core 12 (WFDC12) in the regulation of the inflammatory response in the lung. Thorax 2015; 70: 426–432. doi:10.1136/thoraxjnl-2014-206488
    OpenUrlAbstract/FREE Full Text
  161. ↵
    1. Dorresteijn MJ,
    2. Draisma A,
    3. van der Hoeven JG, et al.
    Lipopolysaccharide-stimulated whole blood cytokine production does not predict the inflammatory response in human endotoxemia. Innate Immun 2010; 16: 248–253. doi:10.1177/1753425909339923
    OpenUrlCrossRefPubMed
  162. ↵
    1. Spiel AO,
    2. Derhaschnig U,
    3. Schwameis M, et al.
    Effects of prasugrel on platelet inhibition during systemic endotoxaemia: a randomized controlled trial. Clin Sci 2012; 123: 591–600. doi:10.1042/CS20120194
    OpenUrl
  163. ↵
    1. Pleiner J,
    2. Mittermayer F,
    3. Schaller G, et al.
    High doses of vitamin C reverse Escherichia coli endotoxin-induced hyporeactivity to acetylcholine in the human forearm. Circulation 2002; 106: 1460–1464. doi:10.1161/01.CIR.0000030184.70207.FF
    OpenUrlAbstract/FREE Full Text
  164. ↵
    1. Leentjens J,
    2. Kox M,
    3. Koch RM, et al.
    Reversal of immunoparalysis in humans in vivo: a double-blind, placebo-controlled, randomized pilot study. Am J Respir Crit Care Med 2012; 186: 838–845. doi:10.1164/rccm.201204-0645OC
    OpenUrlCrossRefPubMedWeb of Science
  165. ↵
    1. Leijte GP,
    2. Kiers D,
    3. van der Heijden W, et al.
    Treatment With acetylsalicylic acid reverses endotoxin tolerance in humans in vivo: a randomized placebo-controlled study. Crit Care Med 2019; 47: 508–516. doi:10.1097/CCM.0000000000003630
    OpenUrl
  166. ↵
    1. Kiers D,
    2. Koch RM,
    3. Hamers L, et al.
    Characterization of a model of systemic inflammation in humans in vivo elicited by continuous infusion of endotoxin. Sci Rep 2017; 7: 1–10. doi:10.1038/srep40149
    OpenUrlCrossRefPubMed
  167. ↵
    1. Taudorf S,
    2. Krabbe KS,
    3. Berg RMG, et al.
    Human models of low-grade inflammation: Bolus versus continuous infusion of endotoxin. Clin Vaccine Immunol 2007; 14: 250–255. doi:10.1128/CVI.00380-06
    OpenUrlAbstract/FREE Full Text
  168. ↵
    1. Ostrowski SR,
    2. Berg RMG,
    3. Windeløv NA, et al.
    Discrepant fibrinolytic response in plasma and whole blood during experimental endotoxemia in healthy volunteers. PLoS One 2013; 8: e59368. doi:10.1371/journal.pone.0059368
    OpenUrlCrossRefPubMed
    1. Berg RMG,
    2. Plovsing RR,
    3. Evans KA, et al.
    Lipopolysaccharide infusion enhances dynamic cerebral autoregulation without affecting cerebral oxygen vasoreactivity in healthy volunteers. Crit Care 2013; 17: R238. doi:10.1186/cc13062
    OpenUrl
    1. Berg RMG,
    2. Plovsing RR,
    3. Ronit A, et al.
    Disassociation of static and dynamic cerebral autoregulatory performance in healthy volunteers after lipopolysaccharide infusion and in patients with sepsis. AJP Regul Integr Comp Physiol 2012; 303: R1127–R1135. doi:10.1152/ajpregu.00242.2012
    OpenUrl
  169. ↵
    1. Geven C,
    2. van Lier D,
    3. Blet A, et al.
    Safety, tolerability and pharmacokinetics/pharmacodynamics of the adrenomedullin antibody adrecizumab in a first-in-human study and during experimental human endotoxaemia in healthy subjects. Br J Clin Pharmacol 2018; 84: 2129–2141. doi:10.1111/bcp.13655
    OpenUrl
  170. ↵
    1. Pillay J,
    2. Ramakers BP,
    3. Kamp VM, et al.
    Functional heterogeneity and differential priming of circulating neutrophils in human experimental endotoxemia. J Leukoc Biol 2010; 88: 211–220. doi:10.1189/jlb.1209793
    OpenUrlCrossRefPubMedWeb of Science
  171. ↵
    1. Thaler B,
    2. Hohensinner PJ,
    3. Krychtiuk KA, et al.
    Differential in vivo activation of monocyte subsets during low-grade inflammation through experimental endotoxemia in humans. Sci Rep 2016; 6: 30162. doi:10.1038/srep30162
    OpenUrlCrossRefPubMed
  172. ↵
    1. Hernandez ML,
    2. Harris B,
    3. Lay JC, et al.
    Comparative airway inflammatory response of normal volunteers to ozone and lipopolysaccharide challenge. Inhal Toxicol 2010; 22: 648–656. doi:10.3109/08958371003610966
    OpenUrlCrossRefPubMedWeb of Science
  173. ↵
    1. Hernandez ML,
    2. Herbst M,
    3. Lay JC, et al.
    Atopic asthmatic patients have reduced airway inflammatory cell recruitment after inhaled endotoxin challenge compared with healthy volunteers. J Allergy Clin Immunol 2012; 130: 869–876. doi:10.1016/j.jaci.2012.05.026
    OpenUrlCrossRefPubMed
  174. ↵
    1. Alexis NE,
    2. Lay JC,
    3. Almond M, et al.
    Acute LPS inhalation in healthy volunteers induces dendritic cell maturation in vivo. J Allergy Clin Immunol 2005; 115: 345–350. doi:10.1016/j.jaci.2004.11.040
    OpenUrlCrossRefPubMedWeb of Science
  175. ↵
    1. Nick JA,
    2. Coldren CD,
    3. Geraci MW, et al.
    Recombinant human activated protein C reduces human endotoxin-induced pulmonary inflammation via inhibition of neutrophil chemotaxis. Blood 2004; 104: 3878–3885. doi:10.1182/blood-2004-06-2140
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
View this article with LENS
Vol 56 Issue 1 Table of Contents
European Respiratory Journal: 56 (1)
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Human lipopolysaccharide models provide mechanistic and therapeutic insights into systemic and pulmonary inflammation
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Human lipopolysaccharide models provide mechanistic and therapeutic insights into systemic and pulmonary inflammation
Daniel Brooks, Laura C. Barr, Sarah Wiscombe, Daniel F. McAuley, A. John Simpson, Anthony J. Rostron
European Respiratory Journal Jul 2020, 56 (1) 1901298; DOI: 10.1183/13993003.01298-2019

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Human lipopolysaccharide models provide mechanistic and therapeutic insights into systemic and pulmonary inflammation
Daniel Brooks, Laura C. Barr, Sarah Wiscombe, Daniel F. McAuley, A. John Simpson, Anthony J. Rostron
European Respiratory Journal Jul 2020, 56 (1) 1901298; DOI: 10.1183/13993003.01298-2019
del.icio.us logo Digg logo Reddit logo Technorati logo Twitter logo CiteULike logo Connotea logo Facebook logo Google logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Abstract
    • Introduction
    • Methods
    • Intravenous LPS model
    • Respiratory LPS models
    • Challenges and limitations of human LPS models
    • Future considerations
    • Shareable PDF
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Patient perceptions of biological therapy for severe asthma
  • Living without eosinophils: evidence from mouse and man
  • Increasing physical activity in severe asthma
Show more Reviews

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERJ

  • Journal information
  • Editorial board
  • Reviewers
  • Press
  • Permissions and reprints
  • Advertising

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • Podcasts
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN:  0903-1936
Online ISSN: 1399-3003

Copyright © 2023 by the European Respiratory Society