Skip to main content
Oxford University Press logoLink to Oxford University Press
. 2023 Dec 8;115(6):1029–1041. doi: 10.1093/jleuko/qiad156

Effect of ethanol exposure on innate immune response in sepsis

Sanjoy Roychowdhury 1, Bishnu Pant 2, Emily Cross 3, Rachel Scheraga 4,5, Vidula Vachharajani 6,7,✉,2
PMCID: PMC11136611  NIHMSID: NIHMS1994072  PMID: 38066660

Abstract

Alcohol use disorder, reported by 1 in 8 critically ill patients, is a risk factor for death in sepsis patients. Sepsis, the leading cause of death, kills over 270,000 patients in the United States alone and remains without targeted therapy. Immune response in sepsis transitions from an early hyperinflammation to persistent inflammation and immunosuppression and multiple organ dysfunction during late sepsis. Innate immunity is the first line of defense against pathogen invasion. Ethanol exposure is known to impair innate and adaptive immune response and bacterial clearance in sepsis patients. Specifically, ethanol exposure is known to modulate every aspect of innate immune response with and without sepsis. Multiple molecular mechanisms are implicated in causing dysregulated immune response in ethanol exposure with sepsis, but targeted treatments have remained elusive. In this article, we outline the effects of ethanol exposure on various innate immune cell types in general and during sepsis.

Keywords: alcohol use disorder, ethanol exposure, innate immune cells, sepsis, septic shock


Ethanol exposure dysregulates innate immune response in sepsis.

1. Introduction

Sepsis, defined as life-threatening organ dysfunction caused by a dysregulated host response to infection, is the leading cause of death in hospitalized patients, killing over 270,000 patients annually in the United States alone.1,2 Currently, no sepsis-specific therapies are available, and the management heavily relies on supportive treatments. Decades of research has suggested a variety of molecular mechanisms; however, currently there are no targeted therapies to treat sepsis. Multiple organ dysfunctions positively correlate with sepsis mortality.3 It is well accepted that the comorbidities modulate the frequency and severity of multiple organ dysfunction in sepsis. Several modifiable and nonmodifiable comorbidities, such as age, obesity, diabetes mellitus, cirrhosis of the liver, underlying immune status, and alcohol use disorder (AUD), are known to affect sepsis mortality.4,5

Specifically, AUD, a modifiable risk factor reported by 1 in 8 critically ill patients, is an independent risk factor for sepsis mortality.6 During the COVID-19 pandemic, the incidence of AUD increased even further.7 Multiple preclinical experimental models have shown that acute and chronic ethanol drinking impairs bacterial clearance. While several mechanisms and molecular targets are identified, therapies to reverse this phenomenon have remained elusive. AUD progressing to liver diseases and/or cirrhosis of liver further adds to the complexity of host immunity in sepsis. Evidence suggests that the comorbidities and complications are more prevalent compared with male counterparts in female patients with alcohol-associated chronic liver disease.8 Increased incidence or worse outcomes in younger females with AUD with sepsis is not evident from existing clinical studies.6,9 However, studies focused on gender and age differences in ethanol with sepsis are in order.

An ideal inflammatory response to an infection would initiate quickly upon pathogen invasion, be of proportionate severity to the infectious insult, have a limited impact on organ function, and resolve in a timely manner back to homeostasis, in which it is poised to respond to the next pathogen invasion. During sepsis, however, the immune response is far from ideal and ios dysregulated with a transition from an early/hyperinflammation to late persistent inflammation and immunosuppression and multiple organ failure.10 Approximately one-third of sepsis mortality occurs during early sepsis, while the majority of the sepsis-related deaths occur late, during persistent inflammation and immunosuppression, without a timely return of homeostasis.11–13

Innate immunity is the first line of defense in any pathogen invasion. Inflammation, a highly conserved process, is a protective mechanism against all pathogenic and nonpathogenic (traumatic, environmental exposures, etc.) insults to the organism. Acute and chronic ethanol exposure dysregulates innate and adaptive immunity in a dose and duration-dependent manner.14 The interplay between the two leads to further dysregulation of the immune response. While others have described the role of adaptive immunity in ethanol with sepsis, in this review we focus on this interplay of ethanol with all aspects of the innate immune response.15–17 Table 1 provides an overview of the effects of ethanol exposure on different innate immune cell types and their potential mechanisms.

Table 1.

Phenotype and functions of different immune cell populations: effect of alcohol.18–29

Cell type Phenotype Function Reference
Neutrophil
Inline graphic
N1:
  • CD54high/CD95high/CD182low

N2:
  • CD54low/CD95low/CD182high

  • High density (HDNs)

  • Low density (LDNs)

Proinflammatory: induces TNFα and IP-10
Higher ability to produce ROS
Induces IL-8, lesser ability to produce ROS
Proinflammatory, phagocytosis intact
Exhausted, phagocytosis impaired
Ohms et al.18
Fridlender et al.19
Ohms et al.18
Cho et al.20
Cho et al.21
Monocyte
Inline graphic
CD14highCD16 low (classical)
CD14highCD16internediate (intermediate)
CD14 lowCD16high (nonclassical)
Increases early (2 h) following alcohol intoxication in human
Decreases TLR4 expression
Enriched population in AH patients
Enhanced expression of CCR2/CD206
Significantly decreased in AH patients.
Janicova et al.22
Dhanda et al.23
Rasmussen et al.24
Macrophage
Inline graphic
CD68+ (human)
F4/80+ (mouse)
Ethanol mutes LPS-stimulated phagocytosis
Ethanol decreases LPS-stimulated phagocytosis and glycolysis
Gandhirajan et al.25
Gandhirajan et al.25
DC
Inline graphic
Myeloid DC: HLA-DRhigh/CD83high Alcohol decreases circulating myeloid DCs
CD11c surface expression decreases
Reduces antigen presentation
Siggins et al.26
ILCs
Inline graphic
ILC1
ILC1: NK1.1CD44high
ILC3
Alcohol does not change the numbers but changes their ability to fight infection
Alcohol increases the population in thymus
Impaired secretion of IL-22
Ruiz-Cortes et al.27
Zhang et al.28
Ruiz-Cortes et al.27
Hendrikx et al.29

AH = alcoholic hepatitis; HDN = high-density neutrophil; LDN = low-density neutrophil; ROS = reactive oxygen species.

2. Ethanol exposure and modulation of pathogen-associated molecular patterns, damage-associated molecular patterns, and pattern recognition receptors

The pathogen response is initiated by a system of pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharides (LPS) (produced by gram-negative bacteria), peptidoglycans and lipoteichoic acid (produced by gram-positive bacteria), lipoproteins, nucleic acids, etc. PAMPs are produced by all microorganisms, not necessarily by just the pathogens.30 Immune cells recognize PAMPs using the pattern recognition receptors (PRRs). The PRRs are expressed on the cell surface and in the cytosol in order to screen for pathogen invasion continuously.31 The PRRs families of toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and C-type lectin receptors (CLRs) detect a variety of pathogens and PAMPs.31,32 PAMPs and their respective TLRs include LPS binding to TLR4, peptidoglycan to TLR2, flagellin binding to TLR5, TLR3 binding to double-stranded RNA ligand, and TLR7/8 binding to RNA fragments, and TLR9 binds to CpG motifs associated with bacterial DNA.33–36 Upon binding, the intracellular TLR signaling and interleukin (IL)-1 receptor domains promote activation of immune response.37 The family of TLRs (2, 4, 7, and 9) trigger receptors expressed on myeloid cells-1, to further carry out the immune response during pathogen invasion in sepsis.38,39 Thrombocytopenia, a known marker of hematological dysfunction in multiple organ failure in sepsis, is known to be associated with increased mortality.40 TLR7 was recently shown to be crucial for platelet activation and platelet-leukocyte aggregates and was implicated along with TLR2 in thrombocytopenia and coagulopathy in experimental model of sepsis.41,42 Historically, sepsis-associated multiple organ failure and death were thought to be solely due to PAMPs.43

Ethanol exposure modulates PRRs in the innate immune cells. Acute ethanol exposure suppresses TLR3 signaling pathway in vivo in mice.44 In addition, evidence also supports that the acute ethanol exposure suppresses interferon (IFN)-related amplification loop and thereby other proinflammatory cytokine and chemokine expressions and increases susceptibility to viral infections.45,46 Evidence implicates acute and chronic ethanol exposure–related interference with actin cytoskeleton that leads to disruption of TLR4-CD14 clustering and decreased proinflammatory cytokine expression in response to LPS.47–49 The suppressive effect of acute ethanol exposure with ethanol levels equivalent to 300 mg/100 mL in male mice on TLR4 with LPS exposure in macrophages is also shown to be via p38-MAPK and extracellularly regulated kinases 1 and 2 (ERK1/2).44,50 TLR4 responsive status is suggested to be crucial for survival during the binge drinking model of acute ethanol exposure with late phase sepsis in female mice with sepsis.51,52 Upon activation, the TLR2 and TLR4 receptors associate with cholesterol-rich domains of the cell membrane. Ethanol exposure affects TLR2 and TLR4 differentially, in that it affects TLR4 but not the TLR2 association with the cholesterol-rich domain and thus differentially affecting membrane localization of these 2 TLRs.53 Others have shown the effect of acute ethanol exposure on TLR7- and TLR9-induced pathways.44,50 In communication between the TLRs, the NLR system of PRRs also modulates pathogen-induced and sterile inflammation, including neuroinflammation, via nuclear factor κB (NFκB) activation.54

The PRR expression is tightly controlled in order to prevent an unrestricted inflammatory response. For example, upon activation by LPS on the cell membrane, TLR4 initiates 2 signaling cascades. The TIRAP–MYD88 induction is followed by the formation of a large protein complex known as “Mydodome,” ultimately culminating into activation of transcription factors like NFκB and transcription of proinflammatory genes. The second pathway involves a complex, which engages TRAM and TRIF, begins an endosome formation and endocytosis of the TLR4, mediated by CD14. Endocytosis of TLR4 essentially terminates MyD88-dependent signaling, preventing unrestricted inflammatory response.32,55,56

It was not until the recognition of the inability of the host immune response to differentiate between “self” and “non-self,” and the “danger model,” that the concept of danger-associated molecular patterns (DAMPs) was tied to infectious and sterile inflammatory response.57 During early sepsis, PAMPs recognized by PRRs stimulate an immune response and release DAMPs into the tissue environment and circulation. Several types of DAMPs such as high-mobility group box 1 (HMGB1), extracellular histones, extracellular cold-inducible RNA binding protein, and heat shock proteins are described in literature.58 HMGB1 is an extensively studied DAMP in sepsis.59–61 HMGB1 release is directly modulated by ethanol exposure via redox pathway, decreased HDAC, and other mechanisms.62–64 HMGB1 is recognized by RAGE, leading to the release of tissue-type plasminogen activator, which ultimately leads to extracellular proteolysis and tissue injury.31 Several innate and adaptive immune cells also secrete HMGB1, which is detected by receptors such as TLR2, TLR4, RAGE, etc. to initiate further local and systemic tissue damage.31 These and other DAMPs known to play a critical role during sepsis, such as mitochondrial DNA, heat shock proteins, interleukins, etc. are reviewed extensively elsewhere.31,43,61

In addition to a direct effect on the PRRs, ethanol exposure modulates release of DAMPs to further impact innate immunity. For example, complement C1q, C3, and C5 are shown to contribute to alcoholic liver injury in female mice with chronic ethanol drinking.65 Chronic alcohol feeding in the mice increases expression of proinflammatory cytokines and DAMPs such as IL-1β, IL-6, and intercellular adhesion molecule-1 via modulation of macrophage chemoattractant protein (macrophage chemoattractant protein 1).66 Similarly, the role of macrophage migration inhibitory factor is known to modulate innate immune response with chronic ethanol exposure in patients.67 The role of ethanol in modulating these and several other DAMPs is extensively reviewed in the literature.34,65,68–70

In addition to the direct effect of ethanol exposure on immune cells, alcoholic hepatitis and cirrhosis are independently associated with increased sepsis-related mortality.6,15 In chronic ethanol exposure, TLR2, TLR4, TLR6, and TLR9 are implicated in hepatic injury via increased oxidative stress.71 Interestingly, TLR4- but not TLR2-dependent mechanism seems to be independent of MyD88.72 Widely used chronic ethanol exposure model, the Lieber–DeCarli liquid diet, uses ethanol with dietary fat, protein, and carbohydrates. Composition of the fatty acids, unsaturated vs saturated, modulates intestinal epithelial integrity, adding further to the ethanol-induced hepatic injury via increased blood endotoxin (LPS) levels.73 Interestingly, dietary insulin feeding attenuates alcohol-induced liver injury via suppression of endotoxin and TLR4 in macrophages by modulation of gut microbiome.74 While out of the scope of this review, a crucial role of ethanol in modulating the gut microbiome and innate immune response thereafter is reviewed extensively elsewhere.75,76

Inflammasomes are large (∼700KD) multiprotein complexes in the immune cells, well known for their ability to control the activation of proteolytic enzyme caspase-1.77 In addition to regulation of proinflammatory cytokines IL-1β and IL-18, Caspase-1 also regulates an inflammatory cell death, pyroptosis.78 Inflammasomes are activated upon detection of DAMPs and PAMPs by using PRRs. A classical inflammasome contains sensor, which is usually a NLR or an AIM2-like (absent melanoma-2-like) receptor, an adaptor protein that is ASC (apoptosis-associated speck-like protein containing CARD), and an effector, caspase pro-caspase-1. The NLRs, such as the NLRPs, assemble their own inflammasomes. The NLRP3 inflammasome is the most studied of the inflammasomes, of particular importance in sepsis.79 Evidence shows that the NLRP3 inflammasome mediates cognitive dysfunction in sepsis via p38-MAPK and ERK phosphorylation.80 Similarly, NLRP3 inflammasome is involved in hepatic dysfunction81 in endotoxemia as well as cardiac82 and renal83 dysfunction, and acute lung injury84 in sepsis.

Ethanol-exposure perturbs NLRP3 inflammasome pathway in innate immune cells. A recent study implicated NLRP3 inflammasome pathway in neutrophil migration and macrophage dysfunction in alcohol-related hepatitis in a rodent model.85 NLRP3 inflammasome pathway was also implicated in alcohol-associated neuroinflammation and atherosclerosis.86,87 The effect of ethanol on inflammasome pathway in sepsis is not very well studied. Interestingly, the NLRP3 inflammasome pathway was implicated in acute ethanol-induced immunosuppression.88 However, the effect of ethanol-induced inhibition of inflammasome activation on individual organ systems needs further evaluation.

3. Ethanol exposure and neutrophil function

Neutrophils play a critical role in responding to pathogen invasion. Although thought to be terminally differentiated cells in the past, a growing body of evidence suggests that neutrophils, like other innate cells, and just as macrophages, also differentiate into subphenotypes.58,89 After production in the bone marrow, mature neutrophils are released into circulation. They are attracted to the site of inflammation/microbial invasion by chemoattractants to exert their proinflammatory and microbicidal activities. Senescent neutrophils return to the bone marrow, where they are engulfed by macrophages and destroyed.58 Like M1/M2 macrophage subtypes, the N1 neutrophils are proinflammatory, and N2 neutrophils are associated with an anti-inflammatory phenotype.58,90 Regulatory neutrophils are thought to be immunosuppressive.91 During sepsis, prolonged lifespan and senescence of neutrophils via antiapoptotic mechanisms are implicated in neutrophil dysfunction.92,93 Classification of neutrophils based on density gradient into high-density neutrophils vs low-density neutrophils (LDNs) shows that the LDN subtype was associated with impaired immune function. An increased LDN population is observed in sepsis patients, and these neutrophils exhibit increased degranulation, decreased phagocytic capacity, and impaired immune function in sepsis.94,95 Moreover, reduced immune function in the LDN subphenotype was found to be associated with increased expression of PD-1 and PD-L1 during sepsis.96

Acute ethanol exposure perturbs neutrophil function. In an experimental model of endotoxemia, acute ethanol exposure muted the neutrophil function by inhibition of macrophage inflammatory protein-2 (MIP-2) and cytokine-induced neutrophil chemoattractant.97 Delayed neutrophil recruitment was implicated with acute ethanol exposure in a streptococcal pneumonia model via muted MIP-2 and cytokine-induced neutrophil chemoattractant.98 Poor neutrophil accumulation with staphylococcal skin infection via disruption of IL-23/IL-17 axis was shown to be a novel mechanism for dysregulation of innate immune response in chronic alcohol abuse subjects, making them susceptible to sepsis.99 Ethanol exposure is shown to increase the LDN population in alcohol-associated hepatitis patients. Interestingly, the LDNs are generated from high-density neutrophils, and after neutrophil extracellular trap (NET) formation in an alcoholic hepatitis model, LDNs exhibit an exhausted phenotype with decreased sensitivity to LPS stimulation.20,21

In a classification of neutrophils based on senescence, they can be divided into (1) nonaged neutrophils with appropriate apoptosis mechanism; (2) aged neutrophils that undergo apoptosis, but over a prolonged period of time; and (3) aged neutrophils that do not undergo apoptosis.58 PD-L1 delays neutrophil apoptosis, and these neutrophils modulate T cell responses during late sepsis.91,100 Evidence also suggests that in face of pathogen invasion, the aged neutrophils do not return to bone marrow. Instead, they migrate to the lungs and serve as the first line of defense as well, and they are destroyed locally by efferocytosis in the tissue.101 Neutrophils can act as antigen-presenting cells to CD4+ T cells in a major histocompatibility complex class II (HLA-DR)–dependent manner, although less efficiently compared with monocytes and dendritic cells (DCs).95,102 LDNs, a mixture of neutrophils at various stages of maturation, are increased in number in sepsis.

Neutrophils defend against an invading pathogen by phagocytosis followed by destruction, using antimicrobial mechanisms such as oxidant damage, or secretion of either an antimicrobial peptide or NETs.61 NETosis is a form of neutrophil death when the granule protein and chromatin bind together to kill the invading bacteria.103 Recently, NETosis was linked with venous thromboembolism and disseminated intravascular coagulation in sepsis and septic shock.104,105 Thus, while conceptually a protective mechanism, excessive NETosis can be harmful and be a target for potential therapeutic use.61,106 NETosis is dysregulated with acute ethanol exposure via CXCL1. In fact, recombinant CXCL1 not only rescues NETosis, but also bacterial clearance and survival in male mice with ethanol with sepsis.107 Thus, harnessing the power of DAMPs such as CXCL1 to regulate NETosis may be a potential therapeutic strategy.

However, the effect of acute or chronic ethanol exposure on neutrophil subphenotypes and their respective functional impact needs further study. Furthermore, the impact of ethanol in the setting of sepsis on neutrophil subphenotypes needs further evaluation. This area of study can lead to novel and targeted therapeutic strategies, much needed for sepsis with or without AUD patients.

4. Ethanol exposure and alteration of monocytes, macrophages, and DCs

4.1. Ethanol exposure and monocytes/macrophages

Monocytes and macrophages play a pivotal role in pathogen clearance during sepsis. Monocytes are generated in the bone marrow, released into the circulation, and differentiate into either macrophages or DCs depending on the tissue microenvironment.108 Circulating monocytes are short-lived compared with tissue-resident macrophages. During early sepsis, monocytes and macrophages exhibit a proinflammatory phenotype characterized by excessive secretion of cytokines and chemokines including tumor necrosis factor α (TNF-α), macrophage chemoattractant protein 1, IL-6, and IFN-γ, and enhanced phagocytosis of the invading pathogens. However, the inflammatory and phagocytic abilities of these cells are dysregulated during late sepsis with persistent inflammation and immunosuppression.109–111

AUD impairs bacterial clearance and dampens infection-induced inflammatory responses by modulating the plasticity and function of monocytes and macrophages. Ethanol exposure also suppresses the overproduction of inflammatory mediators by the monocytes during sepsis.112 The ability of ethanol-exposed immune cells in humanized mice was studied using chimeras of peripheral blood mononuclear cells transferred from healthy volunteers or people with AUD. The humanized mice then received intratracheal Klebsiella pneumoniae or oral Enterococcus faecalis infection. The mice with chimeras from AUD subjects were unable to clear either K. pneumoniae or E. faecalis infection. However, mice receiving chimeras from healthy volunteers showed significantly fewer complications from these infections, suggesting that chronic alcohol abuse dampens immune resistance to bacterial infection.112 A recent report indicates that short-term abstinence after chronic alcohol drinking improves antibacterial immunity against oral E. faecalis infection–induced sepsis in mice.113

Several monocyte subtypes are described in literature. Under normal physiological conditions, majority of the monocytes in the human blood are CD14+/CD16 (classical monocytes), with a lower percentage of the CD14+/CD16+ (intermediate) and CD14+/CD16++ (nonclassical monocytes).114 Following infection, the number of CD14+/CD16++ monocytes increases in circulation. Historically, the monocytes were thought to exclusively exhibit a proinflammatory phenotype with alcohol drinking.115,116 However, recent evidence suggest that chronic alcohol abuse increases the nonclassical monocytes resulting in downregulation of TNF-α, IL-6, HLA-DR, and IL-1β.117,118

Ethanol exposure modulates monocytes to change subtypes over time. During the initial phase of ethanol exposure (acute ethanol exposure), the monocytes are proinflammatory; however, the phenotype gradually turns into an anti-inflammatory during chronic alcohol drinking.22 Chronic alcohol abuse increases the population of a specific subset of these intermediate CD14+/CD16+/CD163 monocytes in circulation, commonly termed as M2b.112 However, recent evidence shows that CD163+ monocytes become highly proinflammatory in nature during sepsis.119 The exact role of M2b/CD163+ monocytes with chronic alcohol use in dampening immunity during ethanol exposure needs further investigation. Monocytes form aggregates with platelets following infection and sepsis.120 These aggregates are linked to the proinflammatory phenotype of the monocytes, but the mechanisms are not well understood. Evidence suggests that in healthy volunteers, while the total leukocyte and granulocyte numbers increase early (within 6 h) of drinking alcohol, the monocyte count increases after 24 h. Interestingly, even if the monocyte numbers increase, the reactive oxygen species–producing monocytes remain low in male and female drinkers; however the phagocytosis capacity decreased only in female drinkers, suggesting deranged overall leukocyte and monocyte function.121

Macrophages, the key immune cell type for pathogen clearance, live longer than monocytes.122 Like monocytes, macrophages show hyperinflammatory phenotype, characterized by increased cytokine production and higher phagocytic ability during the early phase of sepsis, which gradually turns into an anti-inflammatory or immune-suppressive phenotype as identified by reduction in phagocytosis and increased production of anti-inflammatory cytokines. Specifically, ethanol exposure increases the expression of CD206 and IL-10, in addition to decreased expressions of IL-1β, HLA-DR, CD80, and CD86.123 In an alcohol-related hepatitis model, ethanol exposure promotes quantitative expression of Arg1, Mrc1, and IL-10 genes and frequency of CD206+ CD163+ macrophages, suggesting M2 phenotype via KLF4 expression.124 Evidence implicates extracellular vesicles containing miR-27a from ethanol-exposed macrophages in regulating naïve macrophage polarization toward the M2 phenotype.125 Impaired macrophage function is implicated to organ damage and mortality in sepsis. Ethanol exposure mutes proinflammatory response in macrophages.126 Recently, our laboratory has also found that alcohol perturbs the metabolic fitness of the bone marrow–derived macrophages through downregulation of glycolysis and phagocytosis.25 Ethanol exposure induces the cytosolic deacetylase sirtuin 2, which deactivates a rate-limiting glycolytic enzyme, platelet isoform of phosphofructokinase. Increased glycolysis facilitates phagocytosis of the invading pathogens via increased intracellular acidification. Ethanol-driven reduction in glycolysis and its by-product lactate reduced phagocytosis.25

Extracellular ATP plays a critical role in regulating macrophage function and bacterial clearance during sepsis, via binding to its receptors commonly known as P2XRs (inotropic) or P2YRs (metabotropic). Macrophage-specific deficiency in ATP receptor P2X4Rs perturbs bacterial clearance in mice with sepsis.127 Interestingly, alcohol plays a dual role in P2X4R signaling. On one hand, it increases the surface expression of P2XRs,128,129 while it also blocks the activity of these receptors, impairing the downstream signaling.130 More work is needed to dissect the role of P2XRs in macrophages during alcohol-induced sepsis.

4.2. Ethanol exposure and DCs

DCs are the major antigen-presenting cells in the body, critical for pathogen recognition and clearance. DCs, generated from hematopoietic progenitor cells in the bone marrow, are classified as plasmacytoid/circulating DC conventional DCs (cDCs), which are further subdivided into cDC1 and cDC2.131,132 Plasmacytoid DCs are generated from stem cells in the circulation, while the classical DCs are generated from the bone marrow–derived myeloid cells.133 DCs act as a linker between the innate and adaptive immunity. Several studies have shown that the numbers of DCs decrease in both humans and mice during sepsis.134 Ayala et al.135 have shown that the numbers of both splenic and peritoneal DCs decrease 24 h post–cecal ligation puncture–induced polymicrobial sepsis.

Monocytes and DCs also play a critical role in antigen presentation following infection. These cells engulf the invading pathogens, process them by proteolysis in the cytosol, and present them on the plasma membrane in association with HLA-DR to elicit a CD4+ T cell–specific immune response.136 During infection, pathogens try to evade clearance by inhibiting the antigen presentation machinery. Recent studies have demonstrated a correlation between reduced human leukocyte antigen (HLA-DR) expression in monocytes and disease severity (Sequential Organ Failure Assessment scores) in sepsis.137,138 In addition to suppressing the production of inflammatory mediators, alcohol consumption also leads to impairment of antigen presentation and expression of HLA-DR in monocytes and DCs. Monocyte-derived DCs, from acute ethanol-exposed mice, exhibited much lower T cell proliferation capacity compared with controls when challenged with an allo-antigen,139 suggesting that alcohol impairs antigen presentation capacity in DCs.

Mitochondrial DNA is a potent stimulator of DCs. Costimulatory molecules such as CD40, CD80, and CD86 are the most characterized costimulatory molecules expressed in DCs. During sepsis, due to ongoing cell death, the circulating levels of mitochondrial DNA increase. Evidence suggests that bone marrow–derived DCs engulf these circulating mitochondrial DNA, which subsequently inhibit the expression of CD40 and CD86 via increased production of IL-10 and activation of STING pathway leading to immunoparalysis in sepsis.140

Making use of both in vivo and in vitro models, several groups have shown that acute and chronic ethanol exposure not only modulates the monocyte differentiation to DCs,141 but also skews the ability of these cells to produce inflammatory mediators including TNF-α, IL-6, and IL-12 in response to septic insults.134,142 A combined insult of PAMPs (LPS) and DAMPs (poly DA/DT) induces the AIM2 inflammasome in bone marrow–derived DCs, as indicated by increased production of IL-1β and ASC proteins, which is inhibited by alcohol pre-exposure.88 Recent studies have shown that alcohol exposure reduces the expression of CD40 and CD86 in resting bone marrow–derived DCs.142 Collectively, these results suggest that a defective antigen presentation machinery in DCs and monocytes could be one of the decisive factors for making AUD patients more vulnerable to sepsis.

5. Ethanol exposure and innate lymphoid cells

Innate lymphoid cells (ILCs) are innate immune cells play a crucial role during pathogen invasion during sepsis. Ethanol is known to perturb ILC function. The first described prototype are natural killer (NK) cells, and subsequently described lymphoid tissue inducer (LTi) cells have differential functions but are developmentally related.143 Other ILC phenotypes such as NK22 cells, natural cytotoxicity receptor 22 cells or NK receptor positive (NKR+) LTi cells have been previously described. The NK cells secrete IFN-γ whereas the LTi and NKR + LTi-like cells produce IL-17 and/or IL-22.143 IL-17 is crucial for immune response to infection, and IL-17–deficient mice show increased mortality due to decreased neutrophil recruitment to the site of infection.144,145 While IL-22 is not very well studied in sepsis, a recent publication suggested increased IL-22 and IL-17 levels to be significant contributors to sepsis-induced acute lung injury.146 In contrast, treatment with IL-22 was also shown to attenuate sepsis-induced hepatic function via JAK/STAT3 signaling pathway and reduction of hepatocyte apoptosis.147 Thus, the role of IL-22 in sepsis remains controversial.

The 2 populations of NK cells are based mainly on CD56 and CD16 expressions. CD56lo–CD16+ cells are known for pathogen-killing capacity, while CD56hi–CD16– cells secrete large amounts of proinflammatory cytokines.148 NK cells kill pathogens using 2 strategies: (1) the cytotoxic granule pathway in which perforin, a membrane-disrupting protein, and granzymes, the serine proteases secreted by exocytosis, activate apoptosis of target cells via caspase-dependent and -independent pathways149,150; and (2) the death ligand pathway in which the cell death ligand such as FasL engages with death receptors such as Fas/CD95 to induce cell death using classical caspase-induced apoptosis.150

NK cell activation is a double-edged weapon. The debate whether NK cell immune responses are friends or foe of the host is ongoing.149,151,152 In sepsis, NK cells, like other innate immune cells, respond to PAMPs by expression of TLRs.153 The signal transduction follows post-TLR stimulation. The cytokine secretion with other coexisting immune cells acts as potent stimulus for the activation of NK cells at the site of infection. Moreover, IL-12 is a potent IFN-γ inducer post-LPS challenge costimulated by IL-15 and IL-18. IL-10 inhibits IFN-γ production from NK cells.149,154 The IFN-γ stimulus further feeds into the positive feedback loop of more NK cell activation and increased proinflammatory cytokine secretion, leading to tissue damage.149,154 Several studies have established a role of NK cells in the pathogenesis of septic shock.155–157 The roles of IL-12, IL-15, and IFN-γ in NK cell–induced systemic inflammation are well established.158–162

The protective role of NK cells against pathogen invasion is also evident. NK cell depletion not only leads to defective defense against invading pathogen, but also is associated with an immunosuppressive phenotype.163–166 A significant number of studies observed decreased NK cell numbers and function during early sepsis,167,168 while others concluded detrimental role of NK cells in sepsis outcomes.169 Thus, a context-dependent role of NK cells during sepsis remains controversial.

Ethanol exposure is known to influence NK cell activity. The suppressive effect of chronic ethanol exposure on basal NK cell function nearly 3 decades ago.170 The role of ethanol in vitro vs in vivo suggested that the ethanol affects NK cell numbers rather than the activity.171 The role of beta-endorphin in ethanol-induced suppression of NK cells was reported subsequently.172,173 Interestingly, chronic ethanol exposure suppresses basal activity of NK cells by disrupting granzyme B, perforin, and IFNγ.174 The suppressive role of ethanol exposure on NK cell activation in response to poly I:C or IL-2 was shown to be due to decreased production and response to IFN-α in mice, at least partly via induction of endogenous corticosterone.175,176 Low levels of ethanol exposure differentially affect immune cell activity via STAT5. Specifically, low levels of ethanol decrease IL-2–induced activation of NK cells via STAT5, while it increases STAT5 activation in T cells.177

The antifibrotic function of NK cells in fibrotic liver diseases is shown to be inhibited by chronic ethanol consumption by the Gao group. Specifically, they showed that the chronic ethanol exposure abrogates antifibrotic effects of NK cells via downregulation of cytotoxic molecules secreted by NK cells, such as perforin; increased TGF-β secretion by hepatic stellate cells, which decreases NK cell killing activity; increased suppressor of cytokine 1 expression; and increased oxidative stress by hepatocytes and thereby inhibition of IFN-γ signaling.178,179 More recently, NK T 10 cells were implicated in alcoholic steatohepatitis with chronic ethanol consumption.180

Thus, evidence supports that the NK cell activity is perturbed in sepsis and septic shock, although whether NK cells are friend or foe in sepsis is an ongoing debate. Ethanol is known to suppress NK cell activity, as outlined previously and by others.179 The effect of either acute or chronic ethanol exposure on NK cells during acute stress of sepsis is not well understood and needs further investigation.

5.1. Ethanol exposure and innate cell interactions

We describe the role of individual innate immune cell types on sepsis outcomes and the effect of ethanol exposure on each cell type individually in the previous sections. Intercellular communication and interactions are important in face of pathogen invasion, and ethanol is known to affect those as well.26,141,181 For example, as the first responders, neutrophils and monocytes undergo adhesion and extravasation to gain access to subendothelial compartment. In addition to phagocytosis and NETosis, the neutrophils secrete proinflammatory cytokines, chemokines, other DAMPs as described previously to signal and attract innate immune cells such as additional neutrophils/monocytes/DCs etc. Monocytes differentiate into macrophages for effective phagocytosis and pathogen clearance. Circulating DCs, similarly, are attracted to the DAMPs in the interstitial space with an intention to kill pathogen. Acute ethanol exposure dampens while the chronic ethanol exposure increases the proinflammatory milieu, in general as previously described in individual cell types. Acute and chronic ethanol exposures are known to dampen the innate immune response and hence intercellular communications as well via various mechanisms, as summarized in Fig. 1.

Fig. 1.

Fig. 1.

Effect of ethanol exposure on innate immune cell interactions. Ethanol exposure affects interactions between different innate immune cell types in addition to affecting individual cell types. In general, acute ethanol dampens while chronic ethanol exposure increases the proinflammatory and pro-oxidant milieu.

In summary, a poised innate immune response is critical for pathogen invasion in sepsis. Every aspect of innate immunity, including different innate immune cell types and their function, are essential for early response to infection. Ethanol exposure dysregulates innate immune response. Unsurprisingly, the effect of ethanol exposure on innate immune response in sepsis is perturbed, leading to increased morbidity and mortality in patients with sepsis.6 Extensive research shows the effect of ethanol exposure on innate immune cells during sepsis. Several genetic, epigenetic, and metabolic mechanistic targets continue to be discovered in experimental sepsis with ethanol exposure. However, the targeted treatment strategies are elusive. Specific molecular targets in immune response to ethanol with sepsis need further investigation.

Acknowledgments

The table and figure were used with Motifolio software.

Contributor Information

Sanjoy Roychowdhury, Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States.

Bishnu Pant, Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States.

Emily Cross, Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States.

Rachel Scheraga, Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States; Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States.

Vidula Vachharajani, Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States; Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States.

Funding

This work was supported by National Institutes of Health grants R01028763 (V.V.) and R01HL155064 (R.S.).

References

  • 1. Buchman  TG, Simpson  SQ, Sciarretta  KL, Finne  KP, Sowers  N, Collier  M, Chavan  S, Oke  I, Pennini  ME, Santhosh  A, et al.  Sepsis among Medicare beneficiaries: 1. The burdens of sepsis, 2012–2018. Crit Care Med.  2020:48(3):276–288. 10.1097/CCM.0000000000004224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Singer  M, Deutschman  CS, Seymour  CW, Shankar-Hari  M, Annane  D, Bauer  M, Bellomo  R, Bernard  GR, Chiche  JD, Coopersmith  CM, et al.  The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016:315(8):801–810. 10.1001/jama.2016.0287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Aird  WC. The role of the endothelium in severe sepsis and multiple organ dysfunction syndrome. Blood. 2003:101(10):3765–3777. 10.1182/blood-2002-06-1887 [DOI] [PubMed] [Google Scholar]
  • 4. Beck  MK, Jensen  AB, Nielsen  AB, Perner  A, Moseley  PL, Brunak  S. Diagnosis trajectories of prior multi-morbidity predict sepsis mortality. Sci Rep.  2016:6(1):36624. 10.1038/srep36624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Adrie  C, Francais  A, Alvarez-Gonzalez  A, Mounier  R, Azoulay  E, Zahar  JR, Clec'h  C, Goldgran-Toledano  D, Hammer  L, Descorps-Declere  A, et al.  Model for predicting short-term mortality of severe sepsis. Critical Care. 2009:13(3):R72. 10.1186/cc7881 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. O’Brien  JM  Jr, Lu  B, Ali  NA, Martin  GS, Aberegg  SK, Marsh  CB, Lemeshow  S, Douglas  IS. Alcohol dependence is independently associated with sepsis, septic shock, and hospital mortality among adult intensive care unit patients. Crit Care Med.  2007:35(2):345–350. 10.1097/01.CCM.0000254340.91644.B2 [DOI] [PubMed] [Google Scholar]
  • 7. Olaker  VR, Kendall  EK, Wang  CX, Parran  TV, Terebuh  P, Kaelber  DC, Xu  R, Davis  PB. Association of recent sars-cov-2 infection with new-onset alcohol use disorder, January 2020 through january 2022. JAMA Netw Open. 2023:6(2):e2255496. 10.1001/jamanetworkopen.2022.55496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Pemmasani  G, Tremaine  WJ, Suresh Kumar  VC, Aswath  G, Sapkota  B, Karagozian  R, John  S. Sex differences in clinical characteristics and outcomes associated with alcoholic hepatitis. Eur J Gastroenterol Hepatol. 2023:35(10):1192–1196. 10.1097/MEG.0000000000002612 [DOI] [PubMed] [Google Scholar]
  • 9. Hu  C, Wu  T, Ma  S, Huang  W, Xu  Q, Kashani  KB, Hu  B, Li  J. Association of thiamine use with outcomes in patients with sepsis and alcohol use disorder: an analysis of the mimic-iii database. Infect Dis Ther. 2022:11(2):771–786. 10.1007/s40121-022-00603-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Mira  JC, Gentile  LF, Mathias  BJ, Efron  PA, Brakenridge  SC, Mohr  AM, Moore  FA, Moldawer  LL. Sepsis pathophysiology, chronic critical illness, and persistent inflammation-immunosuppression and catabolism syndrome. Crit Care Med.  2016:45(2):253–262. 10.1097/CCM.0000000000002074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Otto  GP, Sossdorf  M, Claus  RA, Rodel  J, Menge  K, Reinhart  K, Bauer  M, Riedemann  NC. The late phase of sepsis is characterized by an increased microbiological burden and death rate. Crit Care. 2011:15(4):R183. 10.1186/cc10332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Boomer  JS, To  K, Chang  KC, Takasu  O, Osborne  DF, Walton  AH, Bricker  TL, Jarman  SD  2nd, Kreisel  D, Krupnick  AS, et al.  Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA. 2011:306(23):2594–2605. 10.1001/jama.2011.1829 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Delano  MJ, Thayer  T, Gabrilovich  S, Kelly-Scumpia  KM, Winfield  RD, Scumpia  PO, Cuenca  AG, Warner  E, Wallet  SM, Wallet  MA, et al.  Sepsis induces early alterations in innate immunity that impact mortality to secondary infection. J Immunol. 2011:186(1):195–202. 10.4049/jimmunol.1002104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Barr  T, Helms  C, Grant  K, Messaoudi  I. Opposing effects of alcohol on the immune system. Prog Neuropsychopharmacol Biol Psychiatry. 2016:65:242–251. 10.1016/j.pnpbp.2015.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Gustot  T, Fernandez  J, Szabo  G, Albillos  A, Louvet  A, Jalan  R, Moreau  R, Moreno  C. Sepsis in alcohol-related liver disease. J Hepatol.  2017:67(5):1031–1050. 10.1016/j.jhep.2017.06.013 [DOI] [PubMed] [Google Scholar]
  • 16. Moss  M. Epidemiology of sepsis: race, sex, and chronic alcohol abuse. Clin Infect Dis. 2005:41(Supplement_7):S490–S497. 10.1086/432003 [DOI] [PubMed] [Google Scholar]
  • 17. Vassallo  GA, Dionisi  T, Tarli  C, Augello  G, Mirijello  A, De Cosmo  S, Gasbarrini  A, Addolorato  G. Alcohol-related liver disease and sepsis. Eur Rev Med Pharmacol Sci.  2021:25(13):4563–4569. 10.26355/eurrev_202107_26249 [DOI] [PubMed] [Google Scholar]
  • 18. Ohms  M, Moller  S, Laskay  T. An attempt to polarize human neutrophils toward n1 and n2 phenotypes in vitro. Front Immunol. 2020:11:532. 10.3389/fimmu.2020.00532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Fridlender  ZG, Sun  J, Kim  S, Kapoor  V, Cheng  G, Ling  L, Worthen  GS, Albelda  SM. Polarization of tumor-associated neutrophil phenotype by tgf-beta: “N1” versus “n2” tan. Cancer Cell. 2009:16(3):183–194. 10.1016/j.ccr.2009.06.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Cho  Y, Bukong  TN, Tornai  D, Babuta  M, Vlachos  IS, Kanata  E, Catalano  D, Szabo  G. Neutrophil extracellular traps contribute to liver damage and increase defective low-density neutrophils in alcohol-associated hepatitis. J Hepatol.  2023:78(1):28–44. 10.1016/j.jhep.2022.08.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Cho  Y, Szabo  G. Two faces of neutrophils in liver disease development and progression. Hepatology. 2021:74(1):503–512. 10.1002/hep.31680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Janicova  A, Haag  F, Xu  B, Garza  AP, Dunay  IR, Neunaber  C, Nowak  AJ, Cavalli  P, Marzi  I, Sturm  R, et al.  Acute alcohol intoxication modulates monocyte subsets and their functions in a time-dependent manner in healthy volunteers. Front Immunol. 2021:12:652488. 10.3389/fimmu.2021.652488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Dhanda  AD, Williams  EL, Yates  E, Lait  PJP, Schewitz-Bowers  LP, Hegazy  D, Cramp  ME, Collins  PL, Lee  RWJ. Intermediate monocytes in acute alcoholic hepatitis are functionally activated and induce il-17 expression in cd4(+) t cells. J Immunol. 2019:203(12):3190–3198. 10.4049/jimmunol.1800742 [DOI] [PubMed] [Google Scholar]
  • 24. Rasmussen  EB, Eriksen  LL, Greisen  SR, Hansen  AL, Carstensen  M, Sandahl  TD, Stoy  S, Kragstrup  TW. Diminished non-classical monocytes in the blood associate with disease severity in alcoholic hepatitis. Clin Exp Gastroenterol. 2021:14:259–267. 10.2147/CEG.S299775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Gandhirajan  A, Roychowdhury  S, Kibler  C, Cross  E, Abraham  S, Bellar  A, Nagy  LE, Scheraga  RG, Vachharajani  V. Sirt2-pfkp interaction dysregulates phagocytosis in macrophages with acute ethanol-exposure. Front Immunol. 2022:13:1079962. 10.3389/fimmu.2022.1079962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Siggins  RW, Bagby  GJ, Molina  P, Dufour  J, Nelson  S, Zhang  P. Alcohol exposure impairs myeloid dendritic cell function in rhesus macaques. Alcohol Clin Exp Res.  2009:33(9):1524–1531. 10.1111/j.1530-0277.2009.00980.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Ruiz-Cortes  K, Villageliu  DN, Samuelson  DR. Innate lymphocytes: role in alcohol-induced immune dysfunction. Front Immunol. 2022:13:934617. 10.3389/fimmu.2022.934617 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Zhang  H, Zhang  F, Zhu  Z, Luong  D, Meadows  GG. Chronic alcohol consumption enhances inkt cell maturation and activation. Toxicol Appl Pharmacol. 2015:282(2):139–150. 10.1016/j.taap.2014.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Hendrikx  T, Duan  Y, Wang  Y, Oh  JH, Alexander  LM, Huang  W, Starkel  P, Ho  SB, Gao  B, Fiehn  O, et al.  Bacteria engineered to produce il-22 in intestine induce expression of reg3g to reduce ethanol-induced liver disease in mice. Gut. 2019:68(8):1504–1515. 10.1136/gutjnl-2018-317232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Medzhitov  R, Janeway  CA  Jr. Decoding the patterns of self and nonself by the innate immune system. Science. 2002:296(5566):298–300. 10.1126/science.1068883 [DOI] [PubMed] [Google Scholar]
  • 31. Rajaee  A, Barnett  R, Cheadle  WG. Pathogen- and danger-associated molecular patterns and the cytokine response in sepsis. Surg Infect (Larchmt).  2018:19(2):107–116. 10.1089/sur.2017.264 [DOI] [PubMed] [Google Scholar]
  • 32. Cao  X. Self-regulation and cross-regulation of pattern-recognition receptor signalling in health and disease. Nat Rev Immunol. 2016:16(1):35–50. 10.1038/nri.2015.8 [DOI] [PubMed] [Google Scholar]
  • 33. de Diego  JL, Gerold  G, Zychlinsky  A. 2007. Sensing, presenting, and regulating pamps. Ernst Schering Found Symp Proc. (3):83–95. [PubMed] [Google Scholar]
  • 34. Pascual  M, Calvo-Rodriguez  M, Nunez  L, Villalobos  C, Urena  J, Guerri  C. Toll-like receptors in neuroinflammation, neurodegeneration, and alcohol-induced brain damage. IUBMB Life. 2021:73(7):900–915. 10.1002/iub.2510 [DOI] [PubMed] [Google Scholar]
  • 35. Lim  CS, Jang  YH, Lee  GY, Han  GM, Jeong  HJ, Kim  JW, Lee  JO. Tlr3 forms a highly organized cluster when bound to a poly(i:C) rna ligand. Nat Commun. 2022:13(1):6876. 10.1038/s41467-022-34602-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Wallach  T, Raden  M, Hinkelmann  L, Brehm  M, Rabsch  D, Weidling  H, Kruger  C, Kettenmann  H, Backofen  R, Lehnardt  S. Distinct sars-cov-2 rna fragments activate toll-like receptors 7 and 8 and induce cytokine release from human macrophages and microglia. Front Immunol. 2022:13:1066456. 10.3389/fimmu.2022.1066456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kopp  E, Medzhitov  R. Recognition of microbial infection by toll-like receptors. Curr Opin Immunol.  2003:15(4):396–401. 10.1016/S0952-7915(03)00080-3 [DOI] [PubMed] [Google Scholar]
  • 38. Prufer  S, Weber  M, Sasca  D, Teschner  D, Wolfel  C, Stein  P, Stassen  M, Schild  H, Radsak  MP. Distinct signaling cascades of trem-1, tlr and nlr in neutrophils and monocytic cells. J Innate Immun. 2014:6(3):339–352. 10.1159/000355892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. van Bremen  T, Dromann  D, Luitjens  K, Dodt  C, Dalhoff  K, Goldmann  T, Schaaf  B. Triggering receptor expressed on myeloid cells-1 (trem-1) on blood neutrophils is associated with cytokine inducibility in human e. Coli sepsis. Diagn Pathol. 2013:8(1):24. 10.1186/1746-1596-8-24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Burunsuzoglu  B, Salturk  C, Karakurt  Z, Ongel  EA, Takir  HB, Kargin  F, Horzum  G, Balci  M, Mocin  O, Adiguzel  N, et al.  Thrombocytopenia: a risk factor of mortality for patients with sepsis in the intensive care unit. Turk Thorac J. 2016:17(1):7–14. 10.5578/ttj.17.1.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Williams  B, Zhu  J, Zou  L, Chao  W. Innate immune tlr7 signaling mediates platelet activation and platelet-leukocyte aggregate formation in murine bacterial sepsis. Platelets. 2022:33(8):1251–1259. 10.1080/09537104.2022.2107627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Williams  B, Neder  J, Cui  P, Suen  A, Tanaka  K, Zou  L, Chao  W. Toll-like receptors 2 and 7 mediate coagulation activation and coagulopathy in murine sepsis. J Thromb Haemost: JTH. 2019:17(10):1683–1693. 10.1111/jth.14543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Gentile  LF, Moldawer  LL. Damps, pamps, and the origins of sirs in bacterial sepsis. Shock. 2013:39(1):113–114. 10.1097/SHK.0b013e318277109c [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Pruett  SB, Zheng  Q, Fan  R, Matthews  K, Schwab  C. Acute exposure to ethanol affects toll-like receptor signaling and subsequent responses: an overview of recent studies. Alcohol. 2004:33(3):235–239. 10.1016/j.alcohol.2004.08.003 [DOI] [PubMed] [Google Scholar]
  • 45. Pruett  SB, Schwab  C, Zheng  Q, Fan  R. Suppression of innate immunity by acute ethanol administration: a global perspective and a new mechanism beginning with inhibition of signaling through tlr3. J Immunol. 2004:173(4):2715–2724. 10.4049/jimmunol.173.4.2715 [DOI] [PubMed] [Google Scholar]
  • 46. Pruett  SB, Fan  R, Zheng  Q. Acute ethanol administration profoundly alters poly i:C-induced cytokine expression in mice by a mechanism that is not dependent on corticosterone. Life Sci. 2003:72(16):1825–1839. 10.1016/S0024-3205(02)02507-9 [DOI] [PubMed] [Google Scholar]
  • 47. Szabo  G, Dolganiuc  A, Dai  Q, Pruett  SB. Tlr4, ethanol, and lipid rafts: a new mechanism of ethanol action with implications for other receptor-mediated effects. J Immunol. 2007:178(3):1243–1249. 10.4049/jimmunol.178.3.1243 [DOI] [PubMed] [Google Scholar]
  • 48. Dai  Q, Pruett  SB. Ethanol suppresses lps-induced toll-like receptor 4 clustering, reorganization of the actin cytoskeleton, and associated tnf-alpha production. Alcohol Clin Exp Res.  2006:30(8):1436–1444. 10.1111/j.1530-0277.2006.00172.x [DOI] [PubMed] [Google Scholar]
  • 49. Dai  Q, Zhang  J, Pruett  SB. Ethanol alters cellular activation and cd14 partitioning in lipid rafts. Biochem Biophys Res Commun.  2005:332(1):37–42. 10.1016/j.bbrc.2005.04.088 [DOI] [PubMed] [Google Scholar]
  • 50. Goral  J, Kovacs  EJ. In vivo ethanol exposure down-regulates tlr2-, tlr4-, and tlr9-mediated macrophage inflammatory response by limiting p38 and erk1/2 activation. J Immunol. 2005:174(1):456–463. 10.4049/jimmunol.174.1.456 [DOI] [PubMed] [Google Scholar]
  • 51. Pruett  SB, Fan  R, Cheng  B, Glover  M, Tan  W, Deng  X. Innate immunity and inflammation in sepsis: mechanisms of suppressed host resistance in mice treated with ethanol in a binge-drinking model. Toxicol Sci. 2010:117(2):314–324. 10.1093/toxsci/kfq215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Bhatty  M, Jan  BL, Tan  W, Pruett  SB, Nanduri  B. Role of acute ethanol exposure and tlr4 in early events of sepsis in a mouse model. Alcohol. 2011:45(8):795–803. 10.1016/j.alcohol.2011.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Dolganiuc  A, Bakis  G, Kodys  K, Mandrekar  P, Szabo  G. Acute ethanol treatment modulates toll-like receptor-4 association with lipid rafts. Alcohol Clin Exp Res.  2006:30(1):76–85. 10.1111/j.1530-0277.2006.00003.x [DOI] [PubMed] [Google Scholar]
  • 54. Montesinos  J, Alfonso-Loeches  S, Guerri  C. Impact of the innate immune response in the actions of ethanol on the central nervous system. Alcohol Clin Exp Res.  2016:40(11):2260–2270. 10.1111/acer.13208 [DOI] [PubMed] [Google Scholar]
  • 55. Ciesielska  A, Matyjek  M, Kwiatkowska  K. Tlr4 and cd14 trafficking and its influence on lps-induced pro-inflammatory signaling. Cell Mol Life Sci. 2021:78(4):1233–1261. 10.1007/s00018-020-03656-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Rajpoot  S, Wary  KK, Ibbott  R, Liu  D, Saqib  U, Thurston  TLM, Baig  MS. Tirap in the mechanism of inflammation. Front Immunol. 2021:12:697588. 10.3389/fimmu.2021.697588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Matzinger  P. The danger model: a renewed sense of self. Science. 2002:296(5566):301–305. 10.1126/science.1071059 [DOI] [PubMed] [Google Scholar]
  • 58. Murao  A, Aziz  M, Wang  P. Neutrophil heterogeneity in sepsis: the role of damage-associated molecular patterns. Shock. 2023:59(2):239–246. 10.1097/SHK.0000000000002019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Coffey  JC, Wang  JH, Kelly  R, Romics  L  Jr, O’Callaghan  A, Fiuza  C, Redmond  HP. Tolerization with blp down-regulates hmgb1 a critical mediator of sepsis-related lethality. J Leukoc Biol.  2007:82(4):906–914. 10.1189/jlb.0806504 [DOI] [PubMed] [Google Scholar]
  • 60. Deng  M, Scott  MJ, Fan  J, Billiar  TR. Location is the key to function: Hmgb1 in sepsis and trauma-induced inflammation. J Leukoc Biol.  2019:106(1):161–169. 10.1002/JLB.3MIR1218-497R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Denning  NL, Aziz  M, Gurien  SD, Wang  P. Damps and nets in sepsis. Front Immunol. 2019:10:2536. 10.3389/fimmu.2019.02536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Frank  MG, Weber  MD, Watkins  LR, Maier  SF. Stress sounds the alarmin: the role of the danger-associated molecular pattern hmgb1 in stress-induced neuroinflammatory priming. Brain Behav Immun. 2015:48:1–7. 10.1016/j.bbi.2015.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Wang  X, Chu  G, Yang  Z, Sun  Y, Zhou  H, Li  M, Shi  J, Tian  B, Zhang  C, Meng  X. Ethanol directly induced hmgb1 release through nox2/nlrp1 inflammasome in neuronal cells. Toxicology. 2015:334:104–110. 10.1016/j.tox.2015.06.006 [DOI] [PubMed] [Google Scholar]
  • 64. Zou  JY, Crews  FT. Release of neuronal hmgb1 by ethanol through decreased hdac activity activates brain neuroimmune signaling. PLoS One. 2014:9(2):e87915. 10.1371/journal.pone.0087915 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Cohen  JI, Roychowdhury  S, McMullen  MR, Stavitsky  AB, Nagy  LE. Complement and alcoholic liver disease: role of c1q in the pathogenesis of ethanol-induced liver injury in mice. Gastroenterology. 2010:139(2):664–674.e661. 10.1053/j.gastro.2010.04.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Mandrekar  P, Ambade  A, Lim  A, Szabo  G, Catalano  D. An essential role for monocyte chemoattractant protein-1 in alcoholic liver injury: regulation of proinflammatory cytokines and hepatic steatosis in mice. Hepatology. 2011:54(6):2185–2197. 10.1002/hep.24599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Kumagi  T, Akbar  F, Horiike  N, Onji  M. Increased serum levels of macrophage migration inhibitory factor in alcoholic liver diseases and their expression in liver tissues. Clin Biochem. 2001:34(3):189–193. 10.1016/S0009-9120(01)00214-4 [DOI] [PubMed] [Google Scholar]
  • 68. Barnes  MA, Roychowdhury  S, Nagy  LE. Innate immunity and cell death in alcoholic liver disease: role of cytochrome p4502e1. Redox Biol.  2014:2:929–935. 10.1016/j.redox.2014.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Chen  Z, Zhang  H, Qu  M, Nan  K, Cao  H, Cata  JP, Chen  W, Miao  C. Review: the emerging role of neutrophil extracellular traps in sepsis and sepsis-associated thrombosis. Front Cell Infect Microbiol. 2021:11:653228. 10.3389/fcimb.2021.653228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Zhao  X, Qi  H, Zhou  J, Xu  S, Gao  Y. Treatment with recombinant interleukin-15 (il-15) increases the number of t cells and natural killer (nk) cells and levels of interferon-gamma (ifn-gamma) in a rat model of sepsis. Med Sci Monit. 2019:25:4450–4456. 10.12659/MSM.914026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Gustot  T, Lemmers  A, Moreno  C, Nagy  N, Quertinmont  E, Nicaise  C, Franchimont  D, Louis  H, Deviere  J, Le Moine  O. Differential liver sensitization to toll-like receptor pathways in mice with alcoholic fatty liver. Hepatology. 2006:43(5):989–1000. 10.1002/hep.21138 [DOI] [PubMed] [Google Scholar]
  • 72. Hritz  I, Mandrekar  P, Velayudham  A, Catalano  D, Dolganiuc  A, Kodys  K, Kurt-Jones  E, Szabo  G. The critical role of toll-like receptor (tlr) 4 in alcoholic liver disease is independent of the common tlr adapter myd88. Hepatology. 2008:48(4):1224–1231. 10.1002/hep.22470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Kirpich  IA, Feng  W, Wang  Y, Liu  Y, Barker  DF, Barve  SS, McClain  CJ. The type of dietary fat modulates intestinal tight junction integrity, gut permeability, and hepatic toll-like receptor expression in a mouse model of alcoholic liver disease. Alcohol Clin Exp Res.  2012:36(5):835–846. 10.1111/j.1530-0277.2011.01673.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Yang  X, He  F, Zhang  Y, Xue  J, Li  K, Zhang  X, Zhu  L, Wang  Z, Wang  H, Yang  S. Inulin ameliorates alcoholic liver disease via suppressing lps-tlr4-mpsi axis and modulating gut microbiota in mice. Alcohol Clin Exp Res.  2019:43(3):411–424. 10.1111/acer.13950 [DOI] [PubMed] [Google Scholar]
  • 75. Tripathi  A, Debelius  J, Brenner  DA, Karin  M, Loomba  R, Schnabl  B, Knight  R. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol. 2018:15(7):397–411. 10.1038/s41575-018-0011-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Hartmann  P, Seebauer  CT, Schnabl  B. Alcoholic liver disease: the gut microbiome and liver cross talk. Alcohol Clin Exp Res.  2015:39(5):763–775. 10.1111/acer.12704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Martinon  F, Burns  K, Tschopp  J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proil-beta. Mol Cell.  2002:10(2):417–426. 10.1016/S1097-2765(02)00599-3 [DOI] [PubMed] [Google Scholar]
  • 78. Rathinam  VA, Fitzgerald  KA. Inflammasome complexes: emerging mechanisms and effector functions. Cell. 2016:165(4):792–800. 10.1016/j.cell.2016.03.046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Shi  X, Tan  S, Tan  S. Nlrp3 inflammasome in sepsis (review). Mol Med Rep. 2021:24(1):514. doi: 10.3892/mmr.2021.12153 [DOI] [PubMed] [Google Scholar]
  • 80. Zhou  R, Yang  X, Li  X, Qu  Y, Huang  Q, Sun  X, Mu  D. Recombinant cc16 inhibits nlrp3/caspase-1-induced pyroptosis through p38 mapk and erk signaling pathways in the brain of a neonatal rat model with sepsis. J Neuroinflammation.  2019:16(1):239. 10.1186/s12974-019-1651-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Tong  Z, Jiang  B, Zhang  L, Liu  Y, Gao  M, Jiang  Y, Li  Y, Lu  Q, Yao  Y, Xiao  X. Hsf-1 is involved in attenuating the release of inflammatory cytokines induced by lps through regulating autophagy. Shock. 2014:41(5):449–453. 10.1097/SHK.0000000000000118 [DOI] [PubMed] [Google Scholar]
  • 82. Zhang  W, Tao  A, Lan  T, Cepinskas  G, Kao  R, Martin  CM, Rui  T. Carbon monoxide releasing molecule-3 improves myocardial function in mice with sepsis by inhibiting nlrp3 inflammasome activation in cardiac fibroblasts. Basic Res Cardiol. 2017:112(2):16. 10.1007/s00395-017-0603-8 [DOI] [PubMed] [Google Scholar]
  • 83. Tanuseputero  SA, Lin  MT, Yeh  SL, Yeh  CL. Intravenous arginine administration downregulates nlrp3 inflammasome activity and attenuates acute kidney injury in mice with polymicrobial sepsis. Mediators Inflamm.  2020:2020:3201635. 10.1155/2020/3201635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Wang  YC, Liu  QX, Zheng  Q, Liu  T, Xu  XE, Liu  XH, Gao  W, Bai  XJ, Li  ZF. Dihydromyricetin alleviates sepsis-induced acute lung injury through inhibiting nlrp3 inflammasome-dependent pyroptosis in mice model. Inflammation. 2019:42(4):1301–1310. 10.1007/s10753-019-00990-7 [DOI] [PubMed] [Google Scholar]
  • 85. Choudhury  A, Bullock  D, Lim  A, Argemi  J, Orning  P, Lien  E, Bataller  R, Mandrekar  P. Inhibition of hsp90 and activation of hsf1 diminish macrophage nlrp3 inflammasome activity in alcohol-associated liver injury. Alcohol Clin Exp Res.  2020:44(6):1300–1311. 10.1111/acer.14338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Lippai  D, Bala  S, Petrasek  J, Csak  T, Levin  I, Kurt-Jones  EA, Szabo  G. Alcohol-induced il-1beta in the brain is mediated by nlrp3/asc inflammasome activation that amplifies neuroinflammation. J Leukoc Biol.  2013:94(1):171–182. 10.1189/jlb.1212659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Abdul-Muneer  PM, Alikunju  S, Mishra  V, Schuetz  H, Szlachetka  AM, Burnham  EL, Haorah  J. Activation of nlrp3 inflammasome by cholesterol crystals in alcohol consumption induces atherosclerotic lesions. Brain Behav Immun. 2017:62:291–305. 10.1016/j.bbi.2017.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Hoyt  LR, Ather  JL, Randall  MJ, DePuccio  DP, Landry  CC, Wewers  MD, Gavrilin  MA, Poynter  ME. Ethanol and other short-chain alcohols inhibit nlrp3 inflammasome activation through protein tyrosine phosphatase stimulation. J Immunol. 2016:197(4):1322–1334. 10.4049/jimmunol.1600406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Xie  X, Shi  Q, Wu  P, Zhang  X, Kambara  H, Su  J, Yu  H, Park  SY, Guo  R, Ren  Q, et al.  Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol.  2020:21(9):1119–1133. 10.1038/s41590-020-0736-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Piccard  H, Muschel  RJ, Opdenakker  G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit Rev Oncol Hematol. 2012:82(3):296–309. 10.1016/j.critrevonc.2011.06.004 [DOI] [PubMed] [Google Scholar]
  • 91. Qi  X, Yu  Y, Sun  R, Huang  J, Liu  L, Yang  Y, Rui  T, Sun  B. Identification and characterization of neutrophil heterogeneity in sepsis. Crit Care. 2021:25(1):50. 10.1186/s13054-021-03481-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Taneja  R, Parodo  J, Jia  SH, Kapus  A, Rotstein  OD, Marshall  JC. Delayed neutrophil apoptosis in sepsis is associated with maintenance of mitochondrial transmembrane potential and reduced caspase-9 activity. Crit Care Med.  2004:32(7):1460–1469. 10.1097/01.CCM.0000129975.26905.77 [DOI] [PubMed] [Google Scholar]
  • 93. Fialkow  L, Fochesatto Filho  L, Bozzetti  MC, Milani  AR, Rodrigues Filho  EM, Ladniuk  RM, Pierozan  P, de Moura  RM, Prolla  JC, Vachon  E, et al.  Neutrophil apoptosis: a marker of disease severity in sepsis and sepsis-induced acute respiratory distress syndrome. Crit Care. 2006:10(6):R155. 10.1186/cc5090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Sun  R, Huang  J, Yang  Y, Liu  L, Shao  Y, Li  L, Sun  B. Dysfunction of low-density neutrophils in peripheral circulation in patients with sepsis. Sci Rep.  2022:12(1):685. 10.1038/s41598-021-04682-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Vono  M, Lin  A, Norrby-Teglund  A, Koup  RA, Liang  F, Lore  K. Neutrophils acquire the capacity for antigen presentation to memory cd4(+) t cells in vitro and ex vivo. Blood. 2017:129(14):1991–2001. 10.1182/blood-2016-10-744441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Patera  AC, Drewry  AM, Chang  K, Beiter  ER, Osborne  D, Hotchkiss  RS. Frontline science: defects in immune function in patients with sepsis are associated with pd-1 or pd-l1 expression and can be restored by antibodies targeting pd-1 or pd-l1. J Leukoc Biol.  2016:100(6):1239–1254. 10.1189/jlb.4HI0616-255R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Zhang  P, Bagby  GJ, Boe  DM, Zhong  Q, Schwarzenberger  P, Kolls  JK, Summer  WR, Nelson  S. Acute alcohol intoxication suppresses the cxc chemokine response during endotoxemia. Alcohol Clin Exp Res.  2002:26(1):65–73. 10.1111/j.1530-0277.2002.tb02433.x [DOI] [PubMed] [Google Scholar]
  • 98. Boe  DM, Nelson  S, Zhang  P, Bagby  GJ. Acute ethanol intoxication suppresses lung chemokine production following infection with streptococcus pneumoniae. J Infect Dis. 2001:184(9):1134–1142. 10.1086/323661 [DOI] [PubMed] [Google Scholar]
  • 99. Parlet  CP, Kavanaugh  JS, Horswill  AR, Schlueter  AJ. Chronic ethanol feeding increases the severity of staphylococcus aureus skin infections by altering local host defenses. J Leukoc Biol.  2015:97(4):769–778. 10.1189/jlb.4A0214-092R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Wang  T, Zhang  X, Liu  Z, Yao  T, Zheng  D, Gan  J, Yu  S, Li  L, Chen  P, Sun  J. Single-cell rna sequencing reveals the sustained immune cell dysfunction in the pathogenesis of sepsis secondary to bacterial pneumonia. Genomics. 2021:113(3):1219–1233. 10.1016/j.ygeno.2021.01.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Uhl  B, Vadlau  Y, Zuchtriegel  G, Nekolla  K, Sharaf  K, Gaertner  F, Massberg  S, Krombach  F, Reichel  CA. Aged neutrophils contribute to the first line of defense in the acute inflammatory response. Blood. 2016:128(19):2327–2337. 10.1182/blood-2016-05-718999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Lin  A, Lore  K. Granulocytes: new members of the antigen-presenting cell family. Front Immunol. 2017:8:1781. 10.3389/fimmu.2017.01781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Brinkmann  V, Reichard  U, Goosmann  C, Fauler  B, Uhlemann  Y, Weiss  DS, Weinrauch  Y, Zychlinsky  A. Neutrophil extracellular traps kill bacteria. Science. 2004:303(5663):1532–1535. 10.1126/science.1092385 [DOI] [PubMed] [Google Scholar]
  • 104. Kimball  AS, Obi  AT, Diaz  JA, Henke  PK. The emerging role of nets in venous thrombosis and immunothrombosis. Front Immunol. 2016:7:236. 10.3389/fimmu.2016.00236 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Delabranche  X, Stiel  L, Severac  F, Galoisy  AC, Mauvieux  L, Zobairi  F, Lavigne  T, Toti  F, Angles-Cano  E, Meziani  F, et al.  Evidence of netosis in septic shock-induced disseminated intravascular coagulation. Shock. 2017:47(3):313–317. 10.1097/SHK.0000000000000719 [DOI] [PubMed] [Google Scholar]
  • 106. Yang  H, Biermann  MH, Brauner  JM, Liu  Y, Zhao  Y, Herrmann  M. New insights into neutrophil extracellular traps: mechanisms of formation and role in inflammation. Front Immunol. 2016:7:302. 10.3389/fimmu.2016.00302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Jin  L, Batra  S, Jeyaseelan  S. Diminished neutrophil extracellular trap (net) formation is a novel innate immune deficiency induced by acute ethanol exposure in polymicrobial sepsis, which can be rescued by cxcl1. PLoS Pathog. 2017:13(9):e1006637. 10.1371/journal.ppat.1006637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Yang  J, Zhang  L, Yu  C, Yang  XF, Wang  H. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res. 2014:2(1):1. 10.1186/2050-7771-2-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Biswas  SK, Lopez-Collazo  E. Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol.  2009:30(10):475–487. 10.1016/j.it.2009.07.009 [DOI] [PubMed] [Google Scholar]
  • 110. Cavaillon  JM, Adrie  C, Fitting  C, Adib-Conquy  M. Endotoxin tolerance: is there a clinical relevance?  J Endotoxin Res. 2003:9(2):101–107. 10.1177/09680519030090020501 [DOI] [PubMed] [Google Scholar]
  • 111. Li  LL, Dai  B, Sun  YH, Zhang  TT. Monocytes undergo functional reprogramming to generate immunosuppression through hif-1alpha signaling pathway in the late phase of sepsis. Mediators Inflamm.  2020:2020:4235909. 10.1155/2020/4235909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Tsuchimoto  Y, Asai  A, Tsuda  Y, Ito  I, Nishiguchi  T, Garcia  MC, Suzuki  S, Kobayashi  M, Higuchi  K, Suzuki  F. M2b monocytes provoke bacterial pneumonia and gut bacteria-associated sepsis in alcoholics. J Immunol. 2015:195(11):5169–5177. 10.4049/jimmunol.1501369 [DOI] [PubMed] [Google Scholar]
  • 113. Kobayashi  M, Asai  A, Ito  I, Suzuki  S, Higuchi  K, Suzuki  F. Short-term alcohol abstinence improves antibacterial defenses of chronic alcohol-consuming mice against gut bacteria-associated sepsis caused by enterococcus faecalis oral infection. Am J Pathol.  2017:187(9):1998–2007. 10.1016/j.ajpath.2017.05.013 [DOI] [PubMed] [Google Scholar]
  • 114. Ziegler-Heitbrock  L, Ancuta  P, Crowe  S, Dalod  M, Grau  V, Hart  DN, Leenen  PJ, Liu  YJ, MacPherson  G, Randolph  GJ, et al.  Nomenclature of monocytes and dendritic cells in blood. Blood. 2010:116(16):e74–e80. 10.1182/blood-2010-02-258558 [DOI] [PubMed] [Google Scholar]
  • 115. Pruett  BS, Pruett  SB. An explanation for the paradoxical induction and suppression of an acute phase response by ethanol. Alcohol. 2006:39(2):105–110. 10.1016/j.alcohol.2006.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Neupane  SP, Skulberg  A, Skulberg  KR, Aass  HC, Bramness  JG. Cytokine changes following acute ethanol intoxication in healthy men: a crossover study. Mediators Inflamm.  2016:2016:3758590. 10.1155/2016/3758590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Szabo  G, Mandrekar  P, Catalano  D. Inhibition of superantigen-induced t cell proliferation and monocyte il-1 beta, tnf-alpha, and il-6 production by acute ethanol treatment. J Leukoc Biol.  1995:58(3):342–350. 10.1002/jlb.58.3.342 [DOI] [PubMed] [Google Scholar]
  • 118. Afshar  M, Richards  S, Mann  D, Cross  A, Smith  GB, Netzer  G, Kovacs  E, Hasday  J. Acute immunomodulatory effects of binge alcohol ingestion. Alcohol. 2015:49(1):57–64. 10.1016/j.alcohol.2014.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Santos  SS, Carmo  AM, Brunialti  MK, Machado  FR, Azevedo  LC, Assuncao  M, Trevelin  SC, Cunha  FQ, Salomao  R. Modulation of monocytes in septic patients: preserved phagocytic activity, increased ros and no generation, and decreased production of inflammatory cytokines. Intensive Care Med Exp. 2016:4(1):5. 10.1186/s40635-016-0078-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Vardon Bounes  F, Memier  V, Marcaud  M, Jacquemin  A, Hamzeh-Cognasse  H, Garcia  C, Series  J, Sie  P, Minville  V, Gratacap  MP, et al.  Platelet activation and prothrombotic properties in a mouse model of peritoneal sepsis. Sci Rep.  2018:8(1):13536. 10.1038/s41598-018-31910-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Haag  F, Janicova  A, Xu  B, Powerski  M, Fachet  M, Bundkirchen  K, Neunaber  C, Marzi  I, Relja  B, Sturm  R. Reduced phagocytosis, ros production and enhanced apoptosis of leukocytes upon alcohol drinking in healthy volunteers. Eur J Trauma Emerg Surg. 2022:48(4):2689–2699. 10.1007/s00068-021-01643-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Malherbe  DC, Messaoudi  I. Transcriptional and epigenetic regulation of monocyte and macrophage dysfunction by chronic alcohol consumption. Front Immunol. 2022:13:911951. 10.3389/fimmu.2022.911951 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. de Castro  LF, de Araujo Mathias  K, Nunes  JV, Galastri  ALB, da Silva  DHL, Longhi  LNA, de Souza Lima Blotta  MH, Mamoni  RL. Ethanol modulates the effector functions of human monocyte-derived macrophages in response to paracoccidioides brasiliensis yeast cells. Med Mycol. 2021:59(8):773–783. 10.1093/mmy/myaa119 [DOI] [PubMed] [Google Scholar]
  • 124. Saha  B, Bala  S, Hosseini  N, Kodys  K, Szabo  G. Kruppel-like factor 4 is a transcriptional regulator of m1/m2 macrophage polarization in alcoholic liver disease. J Leukoc Biol.  2015:97(5):963–973. 10.1189/jlb.4A1014-485R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Saha  B, Momen-Heravi  F, Kodys  K, Szabo  G. Microrna cargo of extracellular vesicles from alcohol-exposed monocytes signals naive monocytes to differentiate into m2 macrophages. J Biol Chem.  2016:291(1):149–159. 10.1074/jbc.M115.694133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Gandhirajan  A, Roychowdhury  S, Kibler  C, Bauer  SR, Nagy  LE, Vachharajani  V. Ethanol exposure attenuates immune response in sepsis via sirtuin 2 expression. Alcohol Clin Exp Res.  2021:45(2):338–350. 10.1111/acer.14542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Csoka  B, Nemeth  ZH, Szabo  I, Davies  DL, Varga  ZV, Paloczi  J, Falzoni  S, Di Virgilio  F, Muramatsu  R, Yamashita  T, et al.  Macrophage p2×4 receptors augment bacterial killing and protect against sepsis. JCI Insight. 2018:3(11):e99431. 10.1172/jci.insight.99431 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Gofman  L, Cenna  JM, Potula  R. P2×4 receptor regulates alcohol-induced responses in microglia. J Neuroimmune Pharmacol. 2014:9(5):668–678. 10.1007/s11481-014-9559-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Asatryan  L, Nam  HW, Lee  MR, Thakkar  MM, Saeed Dar  M, Davies  DL, Choi  DS. Implication of the purinergic system in alcohol use disorders. Alcohol Clin Exp Res.  2011:35(4):584–594. 10.1111/j.1530-0277.2010.01379.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Franklin  KM, Hauser  SR, Lasek  AW, Bell  RL, McBride  WJ. Involvement of purinergic p2×4 receptors in alcohol intake of high-alcohol-drinking (had) rats. Alcohol Clin Exp Res.  2015:39(10):2022–2031. 10.1111/acer.12836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Musumeci  A, Lutz  K, Winheim  E, Krug  AB. What makes a pdc: recent advances in understanding plasmacytoid dc development and heterogeneity. Front Immunol. 2019:10:1222. 10.3389/fimmu.2019.01222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Satpathy  AT, Wu  X, Albring  JC, Murphy  KM. Re(de)fining the dendritic cell lineage. Nat Immunol.  2012:13(12):1145–1154. 10.1038/ni.2467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Fan  X, Liu  Z, Jin  H, Yan  J, Liang  HP. Alterations of dendritic cells in sepsis: featured role in immunoparalysis. Biomed Res Int.  2015:2015:903720. 10.1155/2015/903720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Lau  AH, Abe  M, Thomson  AW. Ethanol affects the generation, cosignaling molecule expression, and function of plasmacytoid and myeloid dendritic cell subsets in vitro and in vivo. J Leukoc Biol.  2006:79(5):941–953. 10.1189/jlb.0905517 [DOI] [PubMed] [Google Scholar]
  • 135. Ding  Y, Chung  CS, Newton  S, Chen  Y, Carlton  S, Albina  JE, Ayala  A. Polymicrobial sepsis induces divergent effects on splenic and peritoneal dendritic cell function in mice. Shock. 2004:22(2):137–144. 10.1097/01.shk.0000131194.80038.3f [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Kotsias  F, Cebrian  I, Alloatti  A. Antigen processing and presentation. Int Rev Cell Mol Biol. 2019:348:69–121. 10.1016/bs.ircmb.2019.07.005 [DOI] [PubMed] [Google Scholar]
  • 137. Papadopoulos  P, Pistiki  A, Theodorakopoulou  M, Christodoulopoulou  T, Damoraki  G, Goukos  D, Briassouli  E, Dimopoulou  I, Armaganidis  A, Nanas  S, et al.  Immunoparalysis: clinical and immunological associations in sirs and severe sepsis patients. Cytokine. 2017:92:83–92. 10.1016/j.cyto.2017.01.012 [DOI] [PubMed] [Google Scholar]
  • 138. Winkler  MS, Rissiek  A, Priefler  M, Schwedhelm  E, Robbe  L, Bauer  A, Zahrte  C, Zoellner  C, Kluge  S, Nierhaus  A. Human leucocyte antigen (hla-dr) gene expression is reduced in sepsis and correlates with impaired tnfalpha response: a diagnostic tool for immunosuppression?  PLoS One. 2017:12(8):e0182427. 10.1371/journal.pone.0182427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Szabo  G, Catalano  D, White  B, Mandrekar  P. Acute alcohol consumption inhibits accessory cell function of monocytes and dendritic cells. Alcohol Clin Exp Res.  2004:28(5):824–828. 10.1097/01.ALC.0000127104.80398.9B [DOI] [PubMed] [Google Scholar]
  • 140. Tu  Q, Li  Y, Zhu  J, Guo  L, Liu  C, Liu  L, Yuan  Y, Zou  Y, Chen  F, Yao  L, et al.  Mitochondrial DNA mediates immunoparalysis of dendritic cells in sepsis via sting signalling. Cell Prolif. 2022:55(12):e13328. 10.1111/cpr.13328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Buttari  B, Profumo  E, Mancinelli  R, Cesta Incani  U, Tosti  ME, Attilia  ML, Ceccanti  M, Rigano  R. Chronic and acute alcohol exposure prevents monocyte-derived dendritic cells from differentiating and maturing. Int J Immunopathol Pharmacol.  2008:21(4):929–939. 10.1177/039463200802100417 [DOI] [PubMed] [Google Scholar]
  • 142. Aloman  C, Gehring  S, Wintermeyer  P, Kuzushita  N, Wands  JR. Chronic ethanol consumption impairs cellular immune responses against hcv ns5 protein due to dendritic cell dysfunction. Gastroenterology. 2007:132(2):698–708. 10.1053/j.gastro.2006.11.016 [DOI] [PubMed] [Google Scholar]
  • 143. Spits  H, Di Santo  JP. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat Immunol.  2011:12(1):21–27. 10.1038/ni.1962 [DOI] [PubMed] [Google Scholar]
  • 144. Freitas  A, Alves-Filho  JC, Victoni  T, Secher  T, Lemos  HP, Sonego  F, Cunha  FQ, Ryffel  B. Il-17 receptor signaling is required to control polymicrobial sepsis. J Immunol. 2009:182(12):7846–7854. 10.4049/jimmunol.0803039 [DOI] [PubMed] [Google Scholar]
  • 145. Bosmann  M, Ward  PA. Therapeutic potential of targeting il-17 and il-23 in sepsis. Clin Transl Med. 2012:1(1):4. 10.1186/2001-1326-1-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Li  G, Zhang  L, Han  N, Zhang  K, Li  H. Increased th17 and th22 cell percentages predict acute lung injury in patients with sepsis. Lung. 2020:198(4):687–693. 10.1007/s00408-020-00362-1 [DOI] [PubMed] [Google Scholar]
  • 147. Liu  EH, Zheng  ZN, Xiao  CX, Liu  X, Lin  XQ. Il-22 relieves sepsis-induced liver injury via activating jak/stat3 signaling pathway. J Biol Regul Homeost Agents. 2020:34(5):1719–1727. 10.23812/20-326-A [DOI] [PubMed] [Google Scholar]
  • 148. Cooper  MA, Fehniger  TA, Caligiuri  MA. The biology of human natural killer-cell subsets. Trends Immunol.  2001:22(11):633–640. 10.1016/S1471-4906(01)02060-9 [DOI] [PubMed] [Google Scholar]
  • 149. Guo  Y, Patil  NK, Luan  L, Bohannon  JK, Sherwood  ER. The biology of natural killer cells during sepsis. Immunology. 2018:153(2):190–202. 10.1111/imm.12854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Smyth  MJ, Cretney  E, Kelly  JM, Westwood  JA, Street  SE, Yagita  H, Takeda  K, van Dommelen  SL, Degli-Esposti  MA, Hayakawa  Y. Activation of nk cell cytotoxicity. Mol Immunol.  2005:42(4):501–510. 10.1016/j.molimm.2004.07.034 [DOI] [PubMed] [Google Scholar]
  • 151. Li  Y, Wang  F, Imani  S, Tao  L, Deng  Y, Cai  Y. Natural killer cells: friend or foe in metabolic diseases?  Front Immunol. 2021:12:614429. 10.3389/fimmu.2021.614429 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Marek  N, Trzonkowski  P. Homeostatic proliferation of nk cells: friend or foe in cellular immunotherapy?  Immunotherapy. 2011:3(6):727–729. 10.2217/imt.11.53 [DOI] [PubMed] [Google Scholar]
  • 153. Souza-Fonseca-Guimaraes  F, Parlato  M, Philippart  F, Misset  B, Cavaillon  JM, Adib-Conquy  M, Captain study g . 2012. Toll-like receptors expression and interferon-gamma production by nk cells in human sepsis. Crit Care. 16(5):R206. 10.1186/cc11838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Varma  TK, Lin  CY, Toliver-Kinsky  TE, Sherwood  ER. Endotoxin-induced gamma interferon production: contributing cell types and key regulatory factors. Clin Diagn Lab Immunol. 2002:9(3):530–543. 10.1128/cdli.9.3.530-543.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Christaki  E, Diza  E, Giamarellos-Bourboulis  EJ, Papadopoulou  N, Pistiki  A, Droggiti  DI, Georgitsi  M, Machova  A, Lambrelli  D, Malisiovas  N, et al.  Nk and nkt cell depletion alters the outcome of experimental pneumococcal pneumonia: relationship with regulation of interferon-gamma production. J Immunol Res.  2015:2015:532717. 10.1155/2015/532717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Badgwell  B, Parihar  R, Magro  C, Dierksheide  J, Russo  T, Carson  WE  3rd. Natural killer cells contribute to the lethality of a murine model of escherichia coli infection. Surgery. 2002:132(2):205–212. 10.1067/msy.2002.125311 [DOI] [PubMed] [Google Scholar]
  • 157. Goldmann  O, Chhatwal  GS, Medina  E. Contribution of natural killer cells to the pathogenesis of septic shock induced by streptococcus pyogenes in mice. J Infect Dis. 2005:191(8):1280–1286. 10.1086/428501 [DOI] [PubMed] [Google Scholar]
  • 158. Heinzel  FP, Rerko  RM, Ling  P, Hakimi  J, Schoenhaut  DS. Interleukin 12 is produced in vivo during endotoxemia and stimulates synthesis of gamma interferon. Infect Immun. 1994:62(10):4244–4249. 10.1128/iai.62.10.4244-4249.1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Jansen  PM, van der Pouw Kraan  TC, de Jong  IW, van Mierlo  G, Wijdenes  J, Chang  AA, Aarden  LA, Taylor  FB  Jr, Hack  CE. Release of interleukin-12 in experimental escherichia coli septic shock in baboons: relation to plasma levels of interleukin-10 and interferon-gamma. Blood. 1996:87(12):5144–5151. 10.1182/blood.V87.12.5144.bloodjournal87125144 [DOI] [PubMed] [Google Scholar]
  • 160. Ito  H, Koide  N, Hassan  F, Islam  S, Tumurkhuu  G, Mori  I, Yoshida  T, Kakumu  S, Moriwaki  H, Yokochi  T. Lethal endotoxic shock using alpha-galactosylceramide sensitization as a new experimental model of septic shock. Lab Invest. 2006:86(3):254–261. 10.1038/labinvest.3700388 [DOI] [PubMed] [Google Scholar]
  • 161. Guo  Y, Luan  L, Patil  NK, Wang  J, Bohannon  JK, Rabacal  W, Fensterheim  BA, Hernandez  A, Sherwood  ER. Il-15 enables septic shock by maintaining nk cell integrity and function. J Immunol. 2017:198(3):1320–1333. 10.4049/jimmunol.1601486 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Guo  Y, Luan  L, Rabacal  W, Bohannon  JK, Fensterheim  BA, Hernandez  A, Sherwood  ER. Il-15 superagonist-mediated immunotoxicity: role of nk cells and ifn-gamma. J Immunol. 2015:195(5):2353–2364. 10.4049/jimmunol.1500300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Pastille  E, Pohlmann  S, Wirsdorfer  F, Reib  A, Flohe  SB. A disturbed interaction with accessory cells upon opportunistic infection with pseudomonas aeruginosa contributes to an impaired ifn-gamma production of nk cells in the lung during sepsis-induced immunosuppression. Innate Immun. 2015:21(2):115–126. 10.1177/1753425913517274 [DOI] [PubMed] [Google Scholar]
  • 164. Hiraki  S, Ono  S, Kinoshita  M, Tsujimoto  H, Takahata  R, Miyazaki  H, Saitoh  D, Seki  S, Hase  K. Neutralization of il-10 restores the downregulation of il-18 receptor on natural killer cells and interferon-gamma production in septic mice, thus leading to an improved survival. Shock. 2012:37(2):177–182. 10.1097/SHK.0b013e31823f18ad [DOI] [PubMed] [Google Scholar]
  • 165. Hall  LJ, Murphy  CT, Hurley  G, Quinlan  A, Shanahan  F, Nally  K, Melgar  S. Natural killer cells protect against mucosal and systemic infection with the enteric pathogen citrobacter rodentium. Infect Immun. 2013:81(2):460–469. 10.1128/IAI.00953-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Nilsson  N, Bremell  T, Tarkowski  A, Carlsten  H. Protective role of nk1.1 + cells in experimental staphylococcus aureus arthritis. Clin Exp Immunol.  1999:117(1):63–69. 10.1046/j.1365-2249.1999.00922.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Chiche  L, Forel  JM, Thomas  G, Farnarier  C, Vely  F, Blery  M, Papazian  L, Vivier  E. The role of natural killer cells in sepsis. J Biomed Biotechnol. 2011:2011:986491. 10.1155/2011/986491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Forel  JM, Chiche  L, Thomas  G, Mancini  J, Farnarier  C, Cognet  C, Guervilly  C, Daumas  A, Vely  F, Xeridat  F, et al.  Phenotype and functions of natural killer cells in critically-ill septic patients. PLoS One. 2012:7(12):e50446. 10.1371/journal.pone.0050446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Giamarellos-Bourboulis  EJ. Natural killer cells in sepsis: detrimental role for final outcome. Crit Care Med.  2014:42(6):1579–1580. 10.1097/CCM.0000000000000352 [DOI] [PubMed] [Google Scholar]
  • 170. Blank  SE, Johansson  JO, Origines  MMT, Meadows  GG. Modulation of nk cell activity by moderate intensity endurance training and chronic ethanol consumption. J Appl Physiol Respir Environ Exerc Physiol.  1992:72(1):8–14. 10.1152/jappl.1992.72.1.8 [DOI] [PubMed] [Google Scholar]
  • 171. Li  F, Cook  RT, Alber  C, Rasmussen  W, Stapleton  JT, Ballas  ZK. Ethanol and natural killer cells. II. Stimulation of human natural killer activity by ethanol in vitro. Alcohol Clin Exp Res.  1997:21(6):981–987. 10.1111/j.1530-0277.1997.tb04241.x [DOI] [PubMed] [Google Scholar]
  • 172. Boyadjieva  N, Dokur  M, Advis  JP, Meadows  GG, Sarkar  DK. Chronic ethanol inhibits nk cell cytolytic activity: role of opioid peptide beta-endorphin. J Immunol. 2001:167(10):5645–5652. 10.4049/jimmunol.167.10.5645 [DOI] [PubMed] [Google Scholar]
  • 173. Boyadjieva  NI, Chaturvedi  K, Poplawski  MM, Sarkar  DK. Opioid antagonist naltrexone disrupts feedback interaction between mu and delta opioid receptors in splenocytes to prevent alcohol inhibition of nk cell function. J Immunol. 2004:173(1):42–49. 10.4049/jimmunol.173.1.42 [DOI] [PubMed] [Google Scholar]
  • 174. Arjona  A, Boyadjieva  N, Sarkar  DK. Circadian rhythms of granzyme b, perforin, ifn-gamma, and nk cell cytolytic activity in the spleen: effects of chronic ethanol. J Immunol. 2004:172(5):2811–2817. 10.4049/jimmunol.172.5.2811 [DOI] [PubMed] [Google Scholar]
  • 175. Collier  SD, Pruett  SB. Mechanisms of suppression of poly i:C-induced activation of nk cells by ethanol. Alcohol. 2000:21(1):87–95. 10.1016/S0741-8329(00)00087-2 [DOI] [PubMed] [Google Scholar]
  • 176. Collier  SD, Wu  WJ, Pruett  SB. Ethanol suppresses nk cell activation by polyinosinic-polycytidylic acid (poly i:c) in female b6c3f1 mice: role of endogenous corticosterone. Alcohol Clin Exp Res.  2000:24(3):291–299. [PubMed] [Google Scholar]
  • 177. Guo  TL, Zhang  LX, Chen  JP, Nguyen  VA, White  K  Jr, Gao  B. Differential stat5 activation and phenotypic marker expression by immune cells following low levels of ethanol consumption in mice. Immunopharmacol Immunotoxicol. 2002:24(1):121–138. 10.1081/IPH-120003408 [DOI] [PubMed] [Google Scholar]
  • 178. Jeong  WI, Park  O, Gao  B. Abrogation of the antifibrotic effects of natural killer cells/interferon-gamma contributes to alcohol acceleration of liver fibrosis. Gastroenterology. 2008:134(1):248–258. 10.1053/j.gastro.2007.09.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Miller  AM, Horiguchi  N, Jeong  WI, Radaeva  S, Gao  B. Molecular mechanisms of alcoholic liver disease: innate immunity and cytokines. Alcohol Clin Exp Res.  2011:35(5):787–793. 10.1111/j.1530-0277.2010.01399.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Cui  K, Yan  G, Zheng  X, Bai  L, Wei  H, Sun  R, Tian  Z. Suppression of natural killer cell activity by regulatory nkt10 cells aggravates alcoholic hepatosteatosis. Front Immunol. 2017:8:1414. 10.3389/fimmu.2017.01414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Bode  C, Bode  JC. Activation of the innate immune system and alcoholic liver disease: effects of ethanol per se or enhanced intestinal translocation of bacterial toxins induced by ethanol?  Alcohol Clin Exp Res.  2005:29(s2):166S–171S. 10.1097/01.alc.0000189280.19073.28 [DOI] [PubMed] [Google Scholar]

Articles from Journal of Leukocyte Biology are provided here courtesy of Oxford University Press

RESOURCES