Skip to main content
Frontiers in Pharmacology logoLink to Frontiers in Pharmacology
. 2015 Oct 23;6:244. doi: 10.3389/fphar.2015.00244

HDL in sepsis – risk factor and therapeutic approach

Emily E Morin 1,2, Ling Guo 3, Anna Schwendeman 1,2,*, Xiang-An Li 3,*
PMCID: PMC4616240  PMID: 26557091

Abstract

High-density lipoprotein (HDL) is a key component of circulating blood and plays essential roles in regulation of vascular endothelial function and immunity. Clinical data demonstrate that HDL levels drop by 40–70% in septic patients, which is associated with a poor prognosis. Experimental studies using Apolipoprotein A-I (ApoAI) null mice showed that HDL deficient mice are susceptible to septic death, and overexpressing ApoAI in mice to increase HDL levels protects against septic death. These clinical and animal studies support our hypothesis that a decrease in HDL level is a risk factor for sepsis, and raising circulating HDL levels may provide an efficient therapy for sepsis. In this review, we discuss the roles of HDL in sepsis and summarize the efforts of using synthetic HDL as a potential therapy for sepsis.

Keywords: high-density lipoprotein (HDL), apolipoprotein A-1 (ApoA1), sepsis, lipopolysaccharide (LPS), sepsis therapy

Introduction

Sepsis is a major health issue in the U. S., claiming over 215,000 lives and causing a financial burden exceeding $17 billion annually (Martin et al., 2003; Riedemann et al., 2003; Angus and van der Poll, 2013). The prognosis for sepsis remains grim, with a mortality rate exceeding 30%, due to poor understanding of the disease and a lack of efficient therapy (Sessler et al., 2004; Lagu et al., 2012; Dellinger et al., 2013).

A major contributor to sepsis mortality is the breakdown in the function of vascular endothelial cells (EC; Aird, 2003; Martin et al., 2003; Riedemann et al., 2003; Deanfield et al., 2007; Angus and van der Poll, 2013). As shown in Figure 1, this breakdown is caused by a cascade of inflammatory events-induced by infections, which includes three major factors/steps: (1) upon infections, bacteria release endotoxin; (2) endotoxin activates immune effector cells to produce inflammatory cytokines and chemokines; (3) inflammatory cytokines and chemokines activates EC, resulting in endothelial dysfunction manifested by vascular leakage, increased leukocyte adhesion, altered vascular tone and a shift in the hemostatic balance toward a pro-coagulant phenotype, which eventually leads to irreversible multi-organ failure and septic death (Aird, 2003; Martin et al., 2003; Riedemann et al., 2003; Deanfield et al., 2007; Angus and van der Poll, 2013). Thus, targeting endothelial dysfunction has been proposed as a potential sepsis therapy. A great challenge is that multiple factors/steps contribute to endothelial dysfunction in sepsis and simply targeting one of the regulatory factors/steps may have limited effect. Indeed, extensive efforts to block one or another component of the inflammatory or coagulation pathways have had little impact on patient survival (Fink and Warren, 2014).

FIGURE 1.

FIGURE 1

Schematic model of vascular endothelial dysfunction in sepsis and high-density lipoprotein (HDL) protection against sepsis.

We believe that targeting an endogenous factor with multi-protective effects against endothelial dysfunction may present a novel approach for sepsis therapy. Emerging evidence suggests that High-density lipoprotein (HDL) is likely such a candidate. In this review, we discuss the roles of HDL in sepsis and summarize the efforts of using synthetic HDL (sHDL) as a potential therapy for sepsis.

Low HDL is a Risk Factor for Sepsis

High-density lipoproteins (HDL) are nanosized protein-lipid particles that circulate throughout the body as a major component of the blood (Navab et al., 2011; Hewing et al., 2012; Zhu and Parks, 2012). Of its physiological functions, HDL is most notably known for its role in cholesterol mobilization and inflammation. HDL, via reverse cholesterol transport, removes cholesterol from peripheral cells and transports it to the liver for excretion in the bile or transport to the adrenals, testes, or ovaries for hormone production (von Eckardstein et al., 2001; Lewis and Rader, 2005; Ohashi et al., 2005; Vickers and Remaley, 2014). In several clinical investigations, plasma HDL levels was shown to be inversely correlated with the occurrence of cardiovascular diseases (CVDs), and many patients with severe CVD have very low levels of circulating HDL (Nofer et al., 2002; Barter et al., 2004; Remaley et al., 2008). Thus, HDL has long been sought after as a possible therapy for the reversal of atherosclerosis and other CVDs. However, HDL is also an important player in inflammation and acute inflammatory disorders. Specifically, HDL has been shown to exert anti-inflammatory properties both in vitro and in vivo (Harris et al., 2002; Barter et al., 2004; Mineo et al., 2006; Argraves and Argraves, 2007; De Nardo et al., 2014; Lüscher et al., 2014) and levels of HDL in patients with inflammatory disorders, such as sepsis, have been proven to be prognostic of clinical outcomes (Gordon, 2004; Riwanto and Landmesser, 2013).

High-density lipoprotein is a strong indicator of both the onset and progression of sepsis. Clinically, HDL levels drop markedly in septic patients, and whether or not these levels rise or continue to fall is often foretelling of their chance of survival (van Leeuwen et al., 2003; Chien et al., 2005; Tsai et al., 2009). In a study of 63 patients, investigators found that those with plasma HDL concentrations exceeding 25 mg/dl at the time of intake had a 100% survival rate (Chien et al., 2005). Additionally, the investigators assessed the power of HDL to predict mortality rate by compartmentalizing patients into two groups: “low” (<20 mg/dl) and “high” (>20 mg/dl) HDL. Using these cutoff values, HDL had a sensitivity of 92%, a specificity of 80%, and an accuracy of 83% for predicting the overall 30-day mortality rate (Chien et al., 2005). The same was done for ApoA1 levels using a cutoff value of 100 mg/dL, however, ApoA1 showed an overall lower predictive value with an accuracy of 73% (Chien et al., 2005), aligning well with other studies showing that HDL, rather than lipid alone, is a better defender against septic shock (Levine et al., 1993).

Other investigations have also been carried out to determine whether low HDL causes septic death. Using ApoA1-null mice as a model for low circulating HDL, it was found that a deficiency in HDL leads to susceptibility to cecal ligation and puncture (CLP)-induced septic death, as well both decreased LPS neutralization and LPS clearance(Guo et al., 2013). Alternatively, increasing HDL levels by over expression of ApoAI improved the survival in both CLP and LPS-induced sepsis models (Li et al., 2008; Guo et al., 2013). Given these clinical and experimental data, we propose that low HDL is a risk factor for sepsis and that targeting HDL may provide an efficient and effective therapy for sepsis.

Mechanisms of HDL Protection Against Sepsis

High-density lipoprotein is a potential multi-protective factor in sepsis. HDL has a broad spectrum of activity, including regulating both immunity and vascular EC functions (Singh et al., 2007; Navab et al., 2011). While most of the existing knowledge of HDL has been acquired in non-sepsis conditions, extensive evidence suggests that HDL likely plays pivotal protective roles in all the steps of endothelial dysfunction (Figure 1), including detoxification of endotoxin, suppression of inflammatory signaling in immune effector cells and inhibition of EC activation.

Detoxification of Endotoxin

Bacterial infections are major causes of sepsis (Vincent et al., 2009). Upon infections, the Gram-negative bacteria release lipopolysaccharides (LPS) which bind to its receptor TLR4 to initiate a downstream signaling cascade. TLR4 binding leads to activation of proinflammatory genes to produce high levels of cytokines such as TNF-α and IL-6, resulting in cell damage (Raetz and Whitfield, 2002; Carmody and Chen, 2007). HDL is well known as a LPS neutralizer (Van Lenten et al., 1986; Flegel et al., 1989; Harris et al., 1990, 1993; Read et al., 1993; Eggesbo et al., 1996; Munford, 2005; Lee et al., 2007; Murch et al., 2007). Most LPS in circulation exist in HDL-bound form (Ulevitch et al., 1979, 1981), and HDL-LPS binding attenuates LPS-TLR4 inflammatory signaling in macrophages (Munford and Dietschy, 1985; Flegel et al., 1989; Baumberger et al., 1991; Emancipator et al., 1992). It is worth noting that simply neutralization of LPS may not provide efficient protection against sepsis which is shown by the failure of anti-LPS monoclonal antibodies in clinical trials (Cohen, 1999). We speculate that the failure of anti-LPS monoclonal antibodies in clinical trials could be attributed to: (i) the apparent inability of antibodies to block LPS-induced cytokine production in human monocytes in vitro (Warren et al., 1993; Cohen, 1999); and (ii) the fact that antibody partially sequester LPS, thus delaying rather than facilitating its clearance (Van Amersfoort et al., 2003). Recent studies including ours suggest that HDL acts together with its receptor, the scavenger receptor BI (SR-BI), to promote LPS clearance (Vishnyakova et al., 2003; Cai et al., 2008; Guo et al., 2009). In vitro, HDL promotes SR-BI-mediated LPS uptake by 4-fold in SR-BI-transfected HEK cells and by twofold in primary hepatocytes (Cai et al., 2008). In vivo, mice deficient in SR-BI or HDL display impaired LPS clearance in LPS or CLP animal models (Cai et al., 2008; Guo et al., 2009, 2013). These findings suggest that HDL neutralizes LPS and promotes LPS clearance via SR-BI-mediated LPS uptake, which presents a more efficient mechanism for LPS detoxification relative to neutralization by anti-LPS antibodies.

Lipoteichoic acid (LTA), released by Gram-positive bacteria, activates the TLR2/6 pathway to generate high levels of inflammatory cytokines, causing cell injury. Similar to LPS, most LTA are associated with HDL in circulation and this HDL-LTA binding neutralizes LTA (Grunfeld et al., 1999; Levels et al., 2001). Given the structural similarity between LPS and LTA, it is likely that HDL neutralizes LTA and promotes LTA clearance via SR-BI-mediated LTA uptake.

Regulating Inflammatory Response in Macrophages

Macrophages are major immune effector cells responsible for inflammatory cytokine production in sepsis (Su, 2002). The inflammatory response in macrophages is required for fighting against infections. However, dysregulation of macrophages produces too many cytokines, leading to vascular endothelial dysfunction and organ injury in sepsis. A body of evidence indicates that HDL is a key modulator of inflammatory response in macrophages (Yvan-Charvet et al., 2008; Zhu et al., 2008; Suzuki et al., 2010; Mineo and Shaul, 2012; Zhu and Parks, 2012; De Nardo et al., 2014): (i) HDL promotes the efflux of free cholesterol from macrophages, resulting in suppression of LPS-induced inflammatory response in macrophages (Mendez et al., 2001; Puff et al., 2005); and ii) HDL upregulates the transcriptional regulator ATF3 which down regulates the expression of inflammatory molecules, resulting in suppression of the inflammatory response in sepsis (De Nardo et al., 2014).

Regulating Endothelial Cell Function

Endothelial cells are activated by LPS and inflammatory cytokines (Aird, 2003; Deanfield et al., 2007; Shapiro et al., 2010). As discussed above, HDL can attenuate EC activation through its roles in promoting LPS detoxification and suppressing inflammatory cytokine production in macrophages. In addition, earlier studies demonstrated that HDL has a variety of activities that modulates EC functions, including: (i) inhibition of adhesion molecule expression stimulated by TNF-α, IL-1β or thrombin (Cockerill et al., 1995, 1999); (ii) activation of eNOS. NO generated by eNOS at small blood vessels is critical for promoting blood supply to small blood vessels and for inhibiting thrombosis in sepsis. Earlier studies including ours demonstrated that HDL activates eNOS to release NO in a SR-BI-dependent manner (Yuhanna et al., 2001; Li et al., 2002; Gong et al., 2003; Mineo et al., 2003); and (iii) prevention of endothelial thrombotic activation by promoting prostacyclin and Cox2 production and suppressing tissue factor and adhesion molecule expression (Ming et al., 2004; Viswambharan et al., 2004; Riwanto and Landmesser, 2013).

In conclusion, HDL likely plays critical roles in promoting LPS/LTA detoxification, suppressing inflammatory response in macrophages and inhibiting EC activation, which may present HDL a multi-protective factor against endothelial dysfunction in sepsis.

Synthetic HDL is a Potential Effective Therapy for Sepsis

Reconstituted or sHDL made from ApoAI protein or ApoAI mimetic peptide presents a new strategy for promoting the biological activity of HDL (Krause and Remaley, 2013). Experimental and clinical investigations, including phase 2 clinical trials for treatment of CVD, have shown that infusion of sHDL raises circulating HDL levels, improves endothelial function and reduces platelet aggregation (Patel et al., 2009; Krause and Remaley, 2013), and with HDL levels at the time of hospitalization being positively correlated with increased survival rates among septic patients (Barlage et al., 2009), it is reasonable that HDL replacement therapy has been a well sought-after area of sepsis research. Not only does HDL confer cardio-protection via maintaining endothelial barrier integrity and reverse cholesterol transport, it is also able to combat inflammation and oxidization, as shown both in vitro and in vivo (Pajkrt et al., 1996). Several studies have been carried out in order to investigate the protective ability of administered HDL against endotoxemia, a few of which are discussed below, and can be found summarized in Table 1.

Table 1.

Experimental high-density lipoprotein (HDL) therapies in animals models of sepsis and their outcomes.

HDL Dose and administration Sepsis model Main findings Reference
18A:Egg PC
(1:2 wt/wt)
sHDL
80 mg/kg; prophylactic tail vein IV infusion Swiss Webster mice;
LPS (salmonella) 10 mg/kg; IP injection; within 15 min of HDL
Three–fourfold increase in 48-h survival rate vs control (p < 0.05) Levine et al., 1993
L-4F Peptide 25 mg/kg, IP, concurrently with lipopolysaccharide (LPS) Sprague-Dawley rats; 10 mg/kg LPS; IP injection Reduction in VCAM-1 expression in excised aortae Gupta et al., 2005
4F Peptide 10 mg/kg; IP injection post-LPS challenge Sprague-Dawley rats; LPS 10 mg/kg or 30 mg/kg; IP injection 10 mg/kg LPS: 4F slowed LPS plasma clearance; reduced hypotension at 6 h;
30 mg/kg LPS: 4F increased plasma HDL levels; increased 24-h survival
Dai et al., 2010
4F Peptide 10 mg/kg, IP, 6 h post-CLP Sprague-Dawley rats; CLP Reduced IL-6; restored CO, right atrial pressure, and plasma volume; improved 2-day survival rate; reversed sepsis-induced changes in lipoprotein profile Zhang et al., 2009
4F Peptide 10 mg/kg; IP injection 6 h post-cecal ligation and puncture (CLP) Wistar Rats; CLP Restored renal, hepatic, and cardiac functions; reduced renal tubule damage; restored expression levels of Slit2, Robo4, and eNOS; increased plasma HDL; improved 4-day survival; no change in MAP Moreira et al., 2014
D-4F Peptide 20 μg daily for 9 days; IP injection C57BL/6J mice; nasally innoculated with 105 PFU influenza virus A/WSN/33 Prevented lymphoid hyperplasia; increased PON activity; prevented drop in core body temperature; suppressed plasma IL-6 levels; increased plasma HDL and inhibited lipoprotein alteration; reduced viral titers by >50% at all time points Van Lenten et al., 2002
ApoA1 10 mg/kg; IP injection 1 h post-LPS challenge Wistar rats; LPS 1 mg/kg (TNF analysis) or 5 mg/kg (survival study); IP injection Reduced plasma TNF-α levels in rats given 1 mg/kg LPS; increased 5-day survival rate from 0 to 90% in rats given 5 mg/kg LPS Imai et al., 2003
ApoA1, human plasma purified 100 mg/kg; IV infusion post-LPS challenge Balb/c mice; LPS 5 mg/kg; IP injection Increased both survival rate and average survival time over 3 days Yan et al., 2006
ApoA1 Milano
ApoA1:Soy PC
(1:3.35 mol/mol)
rHDL
40 mg/kg; prophylactic IV injection Wistar rats; 400 EU/kg Gram-negative bacterial endotoxin; IV injection Increase in HDL-C; improved renal and hepatic function; inhibition of cytokines TNF-α, IL-1β, IL-6; reduced expression of ICAM-1 Zhang et al., 2015
ApoA1:Soy PC (1:200 mol/mol)
rHDL
75 mg/kg ApoA1; prophylactic continuous IV infusion over 25 min NZW rabbits; LPS 25 μg/kg continuous IV infusion over 6 h; start 20 min post-rHDL treatment Complete inhibition of TNF-α; prevented LPS-induced hypotension; reduced metabolic acidosis; no significant effect on serum LPS levels Hubsch et al., 1993
ApoA1:Egg PC
(1:2 w/w)
rHDL
500 mg/kg ApoA1; IV infusion at 0.1 g/kg/hr; split into three doses: 0.3, 0.1, and 0.1 g/kg administered at 0.5, 8, and 16 h post-infection, respectively 2-year old Beagles surgically implanted with E. coli-infected fibrin clot Reduced plasma endotoxin levels; decreased plasma TNF-α; decreased liver function; decreased 2-day survival and average survival time Quezado et al., 1995
CSL-111
ApoA1:Soy PC
(1:150 mol/mol)
rHDL
75 mg/kg ApoA1;
(a) Prophylactic IV infusion over 40 min
(b) Treatment by IV infusion over 20 min, 1 h post-bacterial challenge
NZW Rabbits
(1) Gram (-) Sepsis: 4 × 109 CFU/kg E. coli; IV infusion over 2 h
(2) Gram (+) Sepsis: 2 × 109 CFU/kg; IV infusion over 2 h
(1 a) Prophylactic rHDL: reduced plasma LPS and TNF-α; reduction in metabolic acidosis; no effect on hypotension or blood bacterial levels
(1 b) rHDL Treatment: reduction in LPS after 4 h; reduced metabolic acidosis and creatinine; no effect on blood bacterial counts or TNF-α; no effect on hypotension
(2) No effect in Gram (+) sepsis
Hubsch et al., 1995
CSL-111
ApoA1:Soy PC
(1:150 mol/mol)
rHDL
25 or 50 mg/kg ApoA1; Prophylactic IV infusion over 40 min NZW rabbits; LPS 10 μg/kg; continuous IV infusion over 2 h; start 15 min post-rHDL completion Reduced TNF-α levels and increased TNF-α clearance for both rHDL doses; rHDL 50 mg/kg reduced hypotension at t = 3–4 h; no effect on plasma LPS levels; no effect on blood leukocyte count Casas et al., 1995
CSL-111
ApoA1:Soy PC
(1:150 mol/mol)
rHDL
40 mg/kg; prophylactic IV infusion over 4 h Healthy male volunteers (20–28 years); Endotoxin 4 ng/kg IV bolus; given 3.5 h post-rHDL start Elevated HDL levels; reduced endotoxin-induced clinical symptoms, i.e., chills, myalgia, backache, nausea, and vomiting; reduced plasma cytokine levels of TNF-α, IL-6, and IL-8; inhibited early leukopenia, monopenia, and neutropenia; reduced monocyte CD14 expression Pajkrt et al., 1996

ApoA1

Since ApoA1 is the main protein component of HDL, it makes sense that administering additional ApoA1 protein can increase circulating levels of HDL, avoiding the need to reconstitute with lipid, which adds an additional level of complexity. Administration of naked ApoA1 purified from human plasma has shown to have some beneficial effects in both rat and mouse LPS-induced endotoxemia models. When administered at 10 mg/kg (IP) 1 h post-infection, ApoA1 increased the 5-day survival rate from 0 to 90% in rats given 5 mg/kg LPS (Imai et al., 2003). In a similar model in mice, ApoA1 dosed 1 h post-infection via intravenous (IV) infusion at 100 mg/kg increased both 3-day survival rate and overall survival time versus saline-treated controls (Yan et al., 2006). Additionally, it was found that ApoA1 overexpressing mice were more resistant to infection than those with normal or decreased levels of circulating ApoA1 (Li et al., 2008).

ApoA1 Milano

ApoA1 Milano is a naturally occurring variant of ApoA1 found in a select subset of individuals. Those carrying this mutation, despite having markedly lower levels of circulating HDL, have a much lower risk of developing CVD than their wild-type counterparts (Nissen et al., 2003; Nicholls et al., 2011). It was since developed as an reconstituted HDL (rHDL) therapy by Esperion Therapeutics where it entered a Phase I trial (Nicholls et al., 2011), however, after being licensed to Pfizer it forewent further clinical trials. While a majority of research around ApoA1 Milano is focused around CVD, rHDL using this variant protein (rHDLM) has also been shown to be efficacious against inflammation (Zhang et al., 2015). In a Gram-negative bacterial rat model, rHDLM given prophylactically at 40 mg/kg was effective in suppressing pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 (Zhang et al., 2015). Additionally, rats dosed with rHDLM displayed increased renal and hepatic function as well as a decrease in cardiac tissue damage when compared to saline-treated controls (Zhang et al., 2015).

CSL-111

CSL-111 is a rHDL originally produced by CSL Behring for the intention of treating atherosclerosis, making it through Phase II Clinical Trial before being superseded by CSL-112, CSL Behring’s current investigational rHDL therapeutic (Tardif et al., 2007). Made from purified human ApoA1 and soybean phosphatidylcholine (PC) at a molar ratio of 1:2 protein to lipid, CSL-111 has repeatedly shown efficacy in reducing the burden of LPS-induced endotoxemia both in vitro and in vivo in rabbit and human models (Casas et al., 1995; Hubsch et al., 1995; Pajkrt et al., 1996). In doses ranging from 25 to 75 mg/kg body weight, CSL-111 was able to suppress production of pro-inflammatory cytokines TNF-α, IL-6, and IL-8 (Hubsch et al., 1995; Pajkrt et al., 1996), inhibit sepsis-induced hypotension (Casas et al., 1995; Pajkrt et al., 1996), and markedly decrease the severity of clinical symptoms when administered prophylactically (Pajkrt et al., 1996). Although CSL-111 showed less promise when administered as treatment 1 h post-bacterial challenge, it was still able to reduce the degree metabolic acidosis and improve kidney function over saline controls in rabbit models (Hubsch et al., 1995).

HDL Mimetic Peptides

While HDL therapy may be a feasible solution in the treatment of sepsis, the ability to produce therapeutic quantities of HDL is both a rate- and cost-limiting process in its development. For this reason, the use of ApoA1 mimetic peptides has gained increasing popularity, as they are a cheaper and easier way to make HDL-like particles.

Several mimetic peptides have been synthesized and studied to date, all of which are structurally similar to the amphipathic alpha-helices of native ApoA1 (Navab et al., 2005). Both in vitro and in vivo these peptides are able to bind phospholipids and associate with native HDL particles (Navab et al., 2005, 2011). Functionally, these mimetic peptides are able to reproduce the role of native HDL in their ability to efflux cholesterol (Sethi et al., 2008; Amar et al., 2010), interact with HDL receptors, i.e., ABCA1, ABCG1, and SR-B1 (Sethi et al., 2008), as well as interact with HDL-associated enzymes LCAT and PON (Van Lenten et al., 2002; Chen et al., 2009; Navab et al., 2011), and most relevant to sepsis, the ability to bind and neutralize LPS (Gupta et al., 2005; Remaley et al., 2008).

18A Peptide

One of the first HDL mimetic peptides to be investigated is the 18 amino acid peptide, 18A (DWLKAFYDKVAEKLKEAF). This peptide closely mimics the amphipathic alpha-helical structure of ApoA1, rendering it suitable for sHDL studies. In vivo, prophylactic infusion of 18A sHDL composed of 18A:Egg PC at a 1:2 weight ratio increased the survival rate three–fourfold over saline-treated controls in Swiss Webster mice infected with LPS from salmonella (Levine et al., 1993). Since, 18A peptide has been modified to create 4F peptide, which is discussed below.

4F Peptide

4F is an 18 amino acid peptide (DWFKAFYDKVAEKFKEAF) derived from the sequence of 18A, replacing two lysine residues with phenylalanine (Navab et al., 2005). 4F peptide has been the focus of several studies, and is synthesized using either L- or D-amino acids (L-4F and D-4F, respectively). D-4F is of particular interest to researchers because it can be delivered orally due its resistance to enzymatic degradation (Navab et al., 2005). In a mouse influenza model, D-4F was able to suppress IL-6 production, prevent lymphoid hyperplasia, maintain normal core body temperatures, and reduce viral titers by >50% over the entire course of study when administered intraperitoneally at doses of 20 μg daily (Van Lenten et al., 2002). L-4F has been more widely studied in the context of sepsis, and has been shown to be efficacious in both rat CLP and LPS-induced endotoxemia models at doses as low as 10 mg/kg body weight (Gupta et al., 2005; Zhang et al., 2009; Dai et al., 2010; Moreira et al., 2014). In such studies, L-4F administered by IP injection was shown to block production of cytokines TNF-α and IL-6, reverse sepsis-induced hypotension, prevent organ damage, and restore renal, hepatic, and cardiac function, and increase both survival rate and average survival time in comparison to saline-treated controls (Gupta et al., 2005; Zhang et al., 2009; Dai et al., 2010; Moreira et al., 2014). Most notably, L-4F was given after infection, rather than prophylactically, representing a more clinically relevant application and meriting its further investigation as a potential therapeutic.

Prospectives in sHDL Sepsis Therapy

While previous studies have shown encouraging results, the earlier generation of sHDL and naked ApoAI mimetic peptides suffer from poor purity, short circulation times, contaminations, and toxicity (Quezado et al., 1995; DiPiro et al., 1996; Tardif et al., 2007; Zhang et al., 2009). It is worth noting that the current literature describing sHDL largely focuses on its protein/peptide composition and on its capacity in mediating cholesterol efflux. Considering that sHDL likely plays multi-protective roles in sepsis and the lipid components significantly alter the properties of sHDL, further efforts are required to understand these protective roles and tailor sHDL composition for increased efficacy in sepsis. Extensive efforts have been made to understand the roles of HDL/sHDL in CVD and other chronic inflammatory diseases, and these studies have profoundly improved our understanding about HDL/sHDL. We may take these advantages to further determine the roles of HDL/sHDL in the context of sepsis and develop the new generation of sHDL for sepsis therapy.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Footnotes

Funding. This publication was made possible by Grant Number R01GM113832 (to X-AL and AS) from NIGMS/NIH, AHA 13SDG17230049 (AS), and NIH T32 GM008353 (EM). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIGMS, NIH, or AHA.

References

  1. Aird W. C. (2003). The role of the endothelium in severe sepsis and multiple organ dysfunction syndrome. Blood 101 3765–3777. 10.1182/blood-2002-06-1887 [DOI] [PubMed] [Google Scholar]
  2. Amar M. J. A., D’Souza W., Turner S., Demosky S., Sviridov D., Stonik J., et al. (2010). 5A apolipoprotein mimetic peptide promotes cholesterol efflux and reduces atherosclerosis in mice. J. Pharmacol. Exp. Therapeut. 334 634–641. 10.1124/jpet.110.167890 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Angus D. C., van der Poll T. (2013). Severe sepsis and septic shock. N. Engl. J. Med. 369 840–851. 10.1056/NEJMra1208623 [DOI] [PubMed] [Google Scholar]
  4. Argraves K. M., Argraves W. S. (2007). HDL serves as a S1P signaling platform mediating a multitude of cardiovascular effects. J. Lipid Res. 48 2325–2333. 10.1194/jlr.R700011-JLR200 [DOI] [PubMed] [Google Scholar]
  5. Barlage S., Gnewuch C., Liebisch G., Wolf Z., Audebert F.-X., Glück T., et al. (2009). Changes in HDL-associated apolipoproteins relate to mortality in human sepsis and correlate to monocyte and platelet activation. Intensive Care Med. 35 1877–1885. 10.1007/s00134-009-1609-y [DOI] [PubMed] [Google Scholar]
  6. Barter P. J., Nicholls S., Rye K.-A., Anantharamaiah G. M., Navab M., Fogelman A. M. (2004). Antiinflammatory properties of HDL. Circ. Res. 95 764–772. 10.1161/01.res.0000146094.59640.13 [DOI] [PubMed] [Google Scholar]
  7. Baumberger C., Ulevitch R. J., Dayer J. M. (1991). Modulation of endotoxic activity of lipopolysaccharide by high-density lipoprotein. Pathobiology 59 378–383. 10.1159/000163681 [DOI] [PubMed] [Google Scholar]
  8. Cai L., Ji A., de Beer F. C., Tannock L. R., van der Westhuyzen D. R. (2008). SR-BI protects against endotoxemia in mice through its roles in glucocorticoid production and hepatic clearance. J. Clin. Invest. 118 364–375. 10.1172/JCI31539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Carmody R. J., Chen Y. H. (2007). Nuclear factor-kappaB: activation and regulation during toll-like receptor signaling. Cell. Mol. Immunol. 4 31–41. [PubMed] [Google Scholar]
  10. Casas A. T., Hubsch A. P., Rogers B. C., Doran J. E. (1995). Reconstituted high-density lipoprotein reduces LPS-stimulated TNFα. J. Surg. Res. 59 544–552. 10.1006/jsre.1995.1204 [DOI] [PubMed] [Google Scholar]
  11. Chen X., Burton C., Song X., McNamara L., Langella A., Cianetti S., et al. (2009). An apoA-I mimetic peptide increases LCAT activity in mice through increasing HDL concentration. Int. J. Biol. Sci. 5 489–499. 10.7150/ijbs.5.489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chien J. Y., Jerng J. S., Yu C. J., Yang P. C. (2005). Low serum level of high-density lipoprotein cholesterol is a poor prognostic factor for severe sepsis. Crit. Care Med. 33 1688–1693. 10.1097/01.CCM.0000171183.79525.6B [DOI] [PubMed] [Google Scholar]
  13. Cockerill G. W., Rye K.-A., Gamble J. R., Vadas M. A., Barter P. J. (1995). High-density lipoproteins inhibit cytokine-induced expression of endothelial cell adhesion molecules. Arterioscler. Thromb. Vasc. Biol. 15 1987–1994. 10.1161/01.atv.15.11.1987 [DOI] [PubMed] [Google Scholar]
  14. Cockerill G. W., Saklatvala J., Ridley S. H., Yarwood H., Miller N. E., Oral B., et al. (1999). High-density lipoproteins differentially modulate cytokine-induced expression of e-selectin and cyclooxygenase-2. Arterioscler. Thromb. Vasc. Biol. 19 910–917. 10.1161/01.atv.19.4.910 [DOI] [PubMed] [Google Scholar]
  15. Cohen J. (1999). Adjunctive therapy in sepsis: a critical analysis of the clinical trial programme. Br. Med. Bull. 55 212–225. 10.1258/0007142991902222 [DOI] [PubMed] [Google Scholar]
  16. Dai L., Datta G., Zhang Z., Gupta H., Patel R., Honavar J., et al. (2010). The apolipoprotein A-I mimetic peptide 4F prevents defects in vascular function in endotoxemic rats. J. Lipid Res. 51 2695–2705. 10.1194/jlr.M008086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. De Nardo D., Labzin L. I., Kono H., Seki R., Schmidt S. V., Beyer M., et al. (2014). High-density lipoprotein mediates anti-inflammatory reprogramming of macrophages via the transcriptional regulator ATF3. Nat. Immunol. 15 152–160. 10.1038/ni.2784 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Deanfield J. E., Halcox J. P., Rabelink T. J. (2007). Endothelial function and dysfunction: testing and clinical relevance. Circulation 115 1285–1295. 10.1161/circulationaha.106.652859 [DOI] [PubMed] [Google Scholar]
  19. Dellinger R. P., Levy M. M., Rhodes A., Annane D., Gerlach H., Opal S. M., et al. (2013). Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit. Care Med. 41 580–637. 10.1097/CCM.0b013e31827e83af [DOI] [PubMed] [Google Scholar]
  20. DiPiro J. T., Cue J. I., Richards C. S., Hawkins M. L., Doran J. E., Mansberger A. R. (1996). Pharmacokinetics of reconstituted human high-density lipoprotein in pigs after hemorrhagic shock with resuscitation. Crit. Care Med. 24 440–444. 10.1097/00003246-199603000-00013 [DOI] [PubMed] [Google Scholar]
  21. Eggesbo J. B., Lyberg T., Aspelin T., Hjermann I., Kierulf P. (1996). Different binding of 125I-LPS to plasma proteins from persons with high or low HDL. Scand. J. Clin. Lab. Invest. 56 533–543. 10.3109/00365519609088809 [DOI] [PubMed] [Google Scholar]
  22. Emancipator K., Csako G., Elin R. J. (1992). In vitro inactivation of bacterial endotoxin by human lipoproteins and apolipoproteins. Infect. Immun. 60 596–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Fink M. P., Warren H. S. (2014). Strategies to improve drug development for sepsis. Nat. Rev. Drug Discov. 13 741–758. 10.1038/nrd4368 [DOI] [PubMed] [Google Scholar]
  24. Flegel W. A., Wolpl A., Mannel D. N., Northoff H. (1989). Inhibition of endotoxin-induced activation of human monocytes by human lipoproteins. Infect. Immun. 57 2237–2245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Gong M., Wilson M., Kelly T., Su W., Dressman J., Kincer J., et al. (2003). HDL-associated estradiol stimulates endothelial NO synthase and vasodilation in an SR-BI-dependent manner. J. Clin. Invest. 111 1579–1587. 10.1172/JCI16777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Gordon B. R. (2004). Poor outcomes associated with low lipid and lipoprotein levels. Crit. Care Med. 32 878–879. 10.1097/01.CCM.0000117318.84442.9F [DOI] [PubMed] [Google Scholar]
  27. Grunfeld C., Marshall M., Shigenaga J. K., Moser A. H., Tobias P., Feingold K. R. (1999). Lipoproteins inhibit macrophage activation by lipoteichoic acid. J. Lipid Res. 40 245–252. [PubMed] [Google Scholar]
  28. Guo L., Ai J., Zheng Z., Howatt D. A., Daugherty A., Huang B., et al. (2013). High density lipoprotein protects against polymicrobe-induced sepsis in mice. J. Biol. Chem. 288 17947–17953. 10.1074/jbc.M112.442699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Guo L., Song Z., Li M., Wu Q., Wang D., Feng H., et al. (2009). Scavenger receptor BI protects against septic death through its role in modulating inflammatory response. J. Biol. Chem. 284 19826–19834. 10.1074/jbc.M109.020933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Gupta H., Dai L., Datta G., Garber D. W., Grenett H., Li Y., et al. (2005). Inhibition of lipopolysaccharide-induced inflammatory responses by an apolipoprotein ai mimetic peptide. Circ. Res. 97 236–243. 10.1161/01.res.0000176530.66400.48 [DOI] [PubMed] [Google Scholar]
  31. Harris H. W., Grunfeld C., Feingold K. R., Rapp J. H. (1990). Human very low density lipoproteins and chylomicrons can protect against endotoxin-induced death in mice. J. Clin. Invest. 86 696–702. 10.1172/JCI114765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Harris H. W., Grunfeld C., Feingold K. R., Read T. E., Kane J. P., Jones A. L., et al. (1993). Chylomicrons alter the fate of endotoxin, decreasing tumor necrosis factor release and preventing death. J. Clin. Invest. 91 1028–1034. 10.1172/JCI116259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Harris H. W., Johnson J. A., Wigmore S. J. (2002). Endogenous lipoproteins impact the response to endotoxin in humans. Crit. Care Med. 30 23–31. 10.1097/00003246-200201000-00004 [DOI] [PubMed] [Google Scholar]
  34. Hewing B., Moore K. J., Fisher E. A. (2012). HDL and cardiovascular risk. Circ. Res. 111 1117–1120. 10.1161/circresaha.112.280958 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hubsch A. P., Casas A. T., Doran J. E. (1995). Protective effects of reconstituted high-density lipoprotein in rabbit gram-negative bacteremia models. J. Lab. Clin. Med. 126 548–558. [PubMed] [Google Scholar]
  36. Hubsch A. P., Powell F. S., Lerch P. G., Doran J. E. (1993). A reconstituted, ApoA1 containing lipoprotein reduces tumor necrosis factor release and attenuates shock in endotoxemic rabbits. Circ. Shock 40 14–23. [PubMed] [Google Scholar]
  37. Imai T., Fujita T., Yamazaki Y. (2003). Beneficial effects of apolipoprotein A-I on endotoxemia. Surg. Today 33 684–687. 10.1007/s00595-003-2585-4 [DOI] [PubMed] [Google Scholar]
  38. Krause B. R., Remaley A. T. (2013). Reconstituted HDL for the acute treatment of acute coronary syndrome. Curr. Opin. Lipidol. 24 480–486. 10.1097/MOL.0000000000000020 [DOI] [PubMed] [Google Scholar]
  39. Lagu T., Rothberg M. B., Shieh M. S., Pekow P. S., Steingrub J. S., Lindenauer P. K. (2012). Hospitalizations, costs, and outcomes of severe sepsis in the United States 2003 to 2007. Crit. Care Med. 40 754–761. 10.1097/CCM.0b013e318232db65 [DOI] [PubMed] [Google Scholar]
  40. Lee R. P., Lin N. T., Chao Y. F., Lin C. C., Harn H. J., Chen H. I. (2007). High-density lipoprotein prevents organ damage in endotoxemia. Res. Nurs. Health 30 250–260. 10.1002/nur.20187 [DOI] [PubMed] [Google Scholar]
  41. Levels J. H. M., Abraham P. R., van den Ende A., van Deventer S. J. H. (2001). Distribution and kinetics of lipoprotein-bound endotoxin. Infect. Immun. 69 2821–2828. 10.1128/iai.69.5.2821-2828.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Levine D. M., Parker T. S., Donnelly T. M., Walsh A., Rubin A. L. (1993). In vivo protection against endotoxin by plasma high density lipoprotein. Proc. Natl. Acad. Sci. U.S.A. 90 12040–12044. 10.1073/pnas.90.24.12040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Lewis G. F., Rader D. J. (2005). New insights into the regulation of HDL metabolism and reverse cholesterol transport. Circ. Res. 96 1221–1232. 10.1161/01.RES.0000170946.56981.5c [DOI] [PubMed] [Google Scholar]
  44. Li X. A., Titlow W. B., Jackson B. A., Giltiay N., Nikolova-Karakashian M., Uittenbogaard A., et al. (2002). High density lipoprotein binding to scavenger receptor, Class B, type I activates endothelial nitric-oxide synthase in a ceramide-dependent manner. J. Biol. Chem. 277 11058–11063. 10.1074/jbc.M110985200 [DOI] [PubMed] [Google Scholar]
  45. Li Y., Dong J.-B., Wu M.-P. (2008). Human ApoA-I overexpression diminishes LPS-induced systemic inflammation and multiple organ damage in mice. Eur. J. Pharmacol. 590 417–422. 10.1016/j.ejphar.2008.06.047 [DOI] [PubMed] [Google Scholar]
  46. Lüscher T. F., Landmesser U., von Eckardstein A., Fogelman A. M. (2014). High-density lipoprotein: vascular protective effects, dysfunction, and potential as therapeutic target. Circ. Res. 114 171–182. 10.1161/circresaha.114.300935 [DOI] [PubMed] [Google Scholar]
  47. Martin G. S., Mannino D. M., Eaton S., Moss M. (2003). The epidemiology of sepsis in the United States from 1979 through 2000. N. Engl. J. Med. 348 1546–1554. 10.1056/NEJMoa022139 [DOI] [PubMed] [Google Scholar]
  48. Mendez A. J., Lin G., Wade D. P., Lawn R. M., Oram J. F. (2001). Membrane lipid domains distinct from cholesterol/sphingomyelin-rich rafts are involved in the ABCA1-mediated lipid secretory pathway. J. Biol. Chem. 276 3158–3166. 10.1074/jbc.M007717200 [DOI] [PubMed] [Google Scholar]
  49. Mineo C., Deguchi H., Griffin J. H., Shaul P. W. (2006). Endothelial and antithrombotic actions of HDL. Circ. Res. 98 1352–1364. 10.1161/01.res.0000225982.01988.93 [DOI] [PubMed] [Google Scholar]
  50. Mineo C., Shaul P. W. (2012). Novel biological functions of high-density lipoprotein cholesterol. Circ. Res. 111 1079–1090. 10.1161/circresaha.111.258673 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Mineo C., Yuhanna I. S., Quon M. J., Shaul P. W. (2003). High density lipoprotein-induced endothelial nitric-oxide synthase activation is mediated by Akt and MAP kinases. J. Biol. Chem. 278 9142–9149. 10.1074/jbc.M211394200 [DOI] [PubMed] [Google Scholar]
  52. Ming X. F., Barandier C., Viswambharan H., Kwak B. R., Mach F., Mazzolai L., et al. (2004). Thrombin stimulates human endothelial arginase enzymatic activity via RhoA/ROCK pathway: implications for atherosclerotic endothelial dysfunction. Circulation 110 3708–3714. 10.1161/01.CIR.0000142867.26182.32 [DOI] [PubMed] [Google Scholar]
  53. Moreira R. S., Irigoyen M., Sanches T. R., Volpini R. A., Camara N. O. S., Malheiros D. M., et al. (2014). Apolipoprotein A-I mimetic peptide 4F attenuates kidney injury, heart injury, and endothelial dysfunction in sepsis. Am. J. Physiol. Regul. Integr. Comp. Physiol. 307 R514–R524. 10.1152/ajpregu.00445.2013 [DOI] [PubMed] [Google Scholar]
  54. Munford R. S. (2005). Detoxifying endotoxin: time, place and person. J. Endotoxin. Res. 11 69–84. 10.1179/096805105X35161 [DOI] [PubMed] [Google Scholar]
  55. Munford R. S., Dietschy J. M. (1985). Effects of specific antibodies, hormones, and lipoproteins on bacterial lipopolysaccharides injected into the rat. J. Infect. Dis. 152 177–184. 10.1093/infdis/152.1.177 [DOI] [PubMed] [Google Scholar]
  56. Murch O., Collin M., Hinds C. J., Thiemermann C. (2007). Lipoproteins in inflammation and sepsis. I. Basic science. Intensive Care Med. 33 13–24. 10.1007/s00134-006-0432-y [DOI] [PubMed] [Google Scholar]
  57. Navab M., Anantharamaiah G. M., Reddy S. T., Hama S., Hough G., Grijalva V. R., et al. (2005). Apolipoprotein A-I mimetic peptides. Arterioscler. Thromb. Vasc. Biol. 25 1325–1331. 10.1161/01.atv.0000165694.39518.95 [DOI] [PubMed] [Google Scholar]
  58. Navab M., Reddy S. T., Van Lenten B. J., Fogelman A. M. (2011). HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms. Nat. Rev. Cardiol. 8 222–232. 10.1038/nrcardio.2010.222 [DOI] [PubMed] [Google Scholar]
  59. Nicholls S. J., Uno K., Kataoka Y., Nissen S. E. (2011). ETC-216 for coronary artery disease. Expert Opin. Biol. Ther. 11 387–394. 10.1517/14712598.2011.557061 [DOI] [PubMed] [Google Scholar]
  60. Nissen S. E., Tsunoda T., Tuzcu E. M., Schoenhagen P., Cooper C. J., Yasin M., et al. (2003). Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA 290 2292–2300. 10.1001/jama.290.17.2292 [DOI] [PubMed] [Google Scholar]
  61. Nofer J.-R., Kehrel B., Fobker M., Levkau B., Assmann G., Eckardstein A. V. (2002). HDL and arteriosclerosis: beyond reverse cholesterol transport. Atherosclerosis 161 1–16. 10.1016/S0021-9150(01)00651-7 [DOI] [PubMed] [Google Scholar]
  62. Ohashi R., Mu H., Wang X., Yao Q., Chen C. (2005). Reverse cholesterol transport and cholesterol efflux in atherosclerosis. QJM 98 845–856. 10.1093/qjmed/hci136 [DOI] [PubMed] [Google Scholar]
  63. Pajkrt D., Doran J. E., Koster F., Lerch P. G., Arnet B., van der Poll T., et al. (1996). Antiinflammatory effects of reconstituted high-density lipoprotein during human endotoxemia. J. Exp. Med. 184 1601–1608. 10.1084/jem.184.5.1601 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Patel S., Drew B. G., Nakhla S., Duffy S. J., Murphy A. J., Barter P. J., et al. (2009). Reconstituted high-density lipoprotein increases plasma high-density lipoprotein anti-inflammatory properties and cholesterol efflux capacity in patients with type 2 diabetes. J. Am. Coll. Cardiol. 53 962–971. 10.1016/j.jacc.2008.12.008 [DOI] [PubMed] [Google Scholar]
  65. Puff N., Lamaziere A., Seigneuret M., Trugnan G., Angelova M. I. (2005). HDLs induce raft domain vanishing in heterogeneous giant vesicles. Chem. Phys. Lipids 133 195–202. 10.1016/j.chemphyslip.2004.10.003 [DOI] [PubMed] [Google Scholar]
  66. Quezado Z. M., Natanson C., Banks S. M., Alling D. W., Koev C. A., Danner R. L., et al. (1995). Therapeutic trial of reconstituted human high-density lipoprotein in a canine model of gram-negative septic shock. J. Pharmacol. Exp. Ther. 272 604–611. [PubMed] [Google Scholar]
  67. Raetz C. R., Whitfield C. (2002). Lipopolysaccharide endotoxins. Annu. Rev. Biochem. 71 635–700. 10.1146/annurev.biochem.71.110601.135414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Read T. E., Harris H. W., Grunfeld C., Feingold K. R., Calhoun M. C., Kane J. P., et al. (1993). Chylomicrons enhance endotoxin excretion in bile. Infect. Immun. 61 3496–3502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Remaley A. T., Amar M., Sviridov D. (2008). HDL-replacement therapy: mechanism of action, types of agents and potential clinical indications. Expert Rev. Cardiovasc. Ther. 6 1203–1215. 10.1586/14779072.6.9.1203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Riedemann N. C., Guo R. F., Ward P. A. (2003). The enigma of sepsis. J. Clin. Invest. 112 460–467. 10.1172/JCI200319523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Riwanto M., Landmesser U. (2013). High density lipoproteins and endothelial functions: mechanistic insights and alterations in cardiovascular disease. J. Lipid Res. 54 3227–3243. 10.1194/jlr.R037762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Sessler C. N., Perry J. C., Varney K. L. (2004). Management of severe sepsis and septic shock. Curr. Opin. Crit. Care 10 354–363. 10.1097/01.ccx.0000139363.76068.7b [DOI] [PubMed] [Google Scholar]
  73. Sethi A. A., Stonik J. A., Thomas F., Demosky S. J., Amar M., Neufeld E., et al. (2008). Asymmetry in the lipid affinity of bihelical amphipathic peptides: a structural determinant for the specificity of ABCA1-dependent cholesterol efflux by peptides. J. Biol. Chem. 283 32273–32282. 10.1074/jbc.M804461200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Shapiro N. I., Schuetz P., Yano K., Sorasaki M., Parikh S. M., Jones A. E., et al. (2010). The association of endothelial cell signaling, severity of illness, and organ dysfunction in sepsis. Crit. Care 14:R182 10.1186/cc9290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Singh I. M., Shishehbor M. H., Ansell B. J. (2007). High-density lipoprotein as a therapeutic target: a systematic review. JAMA 298 786–798. 10.1001/jama.298.7.786 [DOI] [PubMed] [Google Scholar]
  76. Su G. L. (2002). Lipopolysaccharides in liver injury: molecular mechanisms of Kupffer cell activation. Am. J. Physiol. Gastrointest. Liver Physiol. 283 G256–G265. 10.1152/ajpgi.00550.2001 [DOI] [PubMed] [Google Scholar]
  77. Suzuki M., Pritchard D. K., Becker L., Hoofnagle A. N., Tanimura N., Bammler T. K., et al. (2010). High-density lipoprotein suppresses the type I interferon response, a family of potent antiviral immunoregulators, in macrophages challenged with lipopolysaccharide. Circulation 122 1919–1927. 10.1161/CIRCULATIONAHA.110.961193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Tardif J. C., Grégoire J., L’Allier P. L., Ibrahim R., Lespérance J., Heinonen T. M., et al. (2007). Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. JAMA 297 1675–1682. 10.1001/jama.297.15.jpc70004 [DOI] [PubMed] [Google Scholar]
  79. Tsai M. H., Peng Y. S., Chen Y. C., Lien J. M., Tian Y. C., Fang J. T., et al. (2009). Low serum concentration of apolipoprotein A-I is an indicator of poor prognosis in cirrhotic patients with severe sepsis. J. Hepatol. 50 906–915. 10.1016/j.jhep.2008.12.024 [DOI] [PubMed] [Google Scholar]
  80. Ulevitch R. J., Johnston A. R., Weinstein D. B. (1979). New function for high density lipoproteins. Their participation in intravascular reactions of bacterial lipopolysaccharides. J. Clin. Invest. 64 1516–1524. 10.1172/JCI109610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Ulevitch R. J., Johnston A. R., Weinstein D. B. (1981). New function for high density lipoproteins. Isolation and characterization of a bacterial lipopolysaccharide-high density lipoprotein complex formed in rabbit plasma. J. Clin. Invest. 67 827–837. 10.1172/JCI110100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Van Amersfoort E. S., Van Berkel T. J., Kuiper J. (2003). Receptors, mediators, and mechanisms involved in bacterial sepsis and septic shock. Clin. Microbiol. Rev. 16 379–414. 10.1128/CMR.16.3.379-414.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. van Leeuwen H. J., Heezius E. C., Dallinga G. M., van Strijp J. A., Verhoef J., van Kessel K. P. (2003). Lipoprotein metabolism in patients with severe sepsis. Crit. Care Med. 31 1359–1366. 10.1097/01.CCM.0000059724.08290.51 [DOI] [PubMed] [Google Scholar]
  84. Van Lenten B. J., Fogelman A. M., Haberland M. E., Edwards P. A. (1986). The role of lipoproteins and receptor-mediated endocytosis in the transport of bacterial lipopolysaccharide. Proc. Natl. Acad. Sci. U.S.A. 83 2704–2708. 10.1073/pnas.83.8.2704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Van Lenten B. J., Wagner A. C., Anantharamaiah G. M., Garber D. W., Fishbein M. C., Adhikary L., et al. (2002). Influenza infection promotes macrophage traffic into arteries of mice that is prevented by D-4F, an apolipoprotein A-I mimetic peptide. Circulation 106 1127–1132. 10.1161/01.cir.0000030182.35880.3e [DOI] [PubMed] [Google Scholar]
  86. Vickers K. C., Remaley A. T. (2014). HDL and cholesterol: life after the divorce? J. Lipid Res. 55 4–12. 10.1194/jlr.R035964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Vincent J. L., Rello J., Marshall J., Silva E., Anzueto A., Martin C. D., et al. (2009). INternational study of the prevalence and outcomes of infection in intensive care units. JAMA 302 2323–2329. 10.1001/jama.2009.1754 [DOI] [PubMed] [Google Scholar]
  88. Vishnyakova T. G., Bocharov A. V., Baranova I. N., Chen Z., Remaley A. T., Csako G., et al. (2003). Binding and internalization of lipopolysaccharide by Cla-1, a human orthologue of rodent scavenger receptor B1. J. Biol. Chem. 278 22771–22780. 10.1074/jbc.M211032200 [DOI] [PubMed] [Google Scholar]
  89. Viswambharan H., Ming X. F., Zhu S., Hubsch A., Lerch P., Vergeres G., et al. (2004). Reconstituted high-density lipoprotein inhibits thrombin-induced endothelial tissue factor expression through inhibition of RhoA and stimulation of phosphatidylinositol 3-kinase but not Akt/endothelial nitric oxide synthase. Circ. Res. 94 918–925. 10.1161/01.RES.0000124302.20396.B7 [DOI] [PubMed] [Google Scholar]
  90. von Eckardstein A., Nofer J.-R., Assmann G. (2001). High density lipoproteins and arteriosclerosis: role of cholesterol efflux and reverse cholesterol transport. Arterioscler. Thromb. Vasc. Biol. 21 13–27. 10.1161/01.atv.21.1.13 [DOI] [PubMed] [Google Scholar]
  91. Warren H. S., Amato S. F., Fitting C., Black K. M., Loiselle P. M., Pasternack M. S., et al. (1993). Assessment of ability of murine and human anti-lipid A monoclonal antibodies to bind and neutralize lipopolysaccharide. J. Exp. Med. 177 89–97. 10.1084/jem.177.1.89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Yan Y.-J., Li Y., Lou B., Wu M.-P. (2006). Beneficial effects of ApoA-I on LPS-induced acute lung injury and endotoxemia in mice. Life Sci. 79 210–215. 10.1016/j.lfs.2006.02.011 [DOI] [PubMed] [Google Scholar]
  93. Yuhanna I. S., Zhu Y., Cox B. E., Hahner L. D., Osborne-Lawrence S., Lu P., et al. (2001). High-density lipoprotein binding to scavenger receptor-BI activates endothelial nitric oxide synthase. Nat. Med. 7 853–857. 10.1038/89986 [DOI] [PubMed] [Google Scholar]
  94. Yvan-Charvet L., Welch C., Pagler T. A., Ranalletta M., Lamkanfi M., Han S., et al. (2008). Increased inflammatory gene expression in ABC transporter-deficient macrophages: free cholesterol accumulation, increased signaling via toll-like receptors, and neutrophil infiltration of atherosclerotic lesions. Circulation 118 1837–1847. 10.1161/CIRCULATIONAHA.108.793869 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Zhang X., Wang L., Chen B. (2015). Recombinant HDL (Milano) protects endotoxin-challenged rats from multiple organ injury and dysfunction. Biol. Chem. 396 53–60. 10.1515/hsz-2014-0188 [DOI] [PubMed] [Google Scholar]
  96. Zhang Z., Datta G., Zhang Y., Miller A. P., Mochon P., Chen Y.-F., et al. (2009). Apolipoprotein A-I mimetic peptide treatment inhibits inflammatory responses and improves survival in septic rats. Am. J. Physiol. Heart Circ. Physiol. 297 H866–H873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Zhu X., Lee J. Y., Timmins J. M., Brown J. M., Boudyguina E., Mulya A., et al. (2008). Increased cellular free cholesterol in macrophage-specific Abca1 knock-out mice enhances pro-inflammatory response of macrophages. J. Biol. Chem. 283 22930–22941. 10.1074/jbc.M801408200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Zhu X., Parks J. S. (2012). New roles of HDL in inflammation and hematopoiesis. Annu. Rev. Nutr. 32 161–182. 10.1146/annurev-nutr-071811-150709 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Frontiers in Pharmacology are provided here courtesy of Frontiers Media SA

RESOURCES