Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jan 1.
Published in final edited form as: Prog Lipid Res. 2010 Sep 29;50(1):75–88. doi: 10.1016/j.plipres.2010.09.002

Resolvins: Natural Agonists for Resolution of Pulmonary Inflammation

Mohib Uddin 1, Bruce D Levy 1
PMCID: PMC3012139  NIHMSID: NIHMS249335  PMID: 20887750

Abstract

Inappropriate or excessive pulmonary inflammation can contribute to chronic lung diseases. In health, the resolution of inflammation is an active process that terminates inflammatory responses. The recent identification of endogenous lipid-derived mediators of resolution has provided a window to explore the pathobiology of inflammatory disease and structural templates for the design of novel pro-resolving therapeutics. Resolvins (resolution-phase interaction products) are a family of pro-resolving mediators that are enzymatically generated from essential omega-3 polyunsaturated fatty acids. Two molecular series of resolvins have been characterized, namely E- and D-series resolvins which possess distinct structural, biochemical and pharmacological properties. Acting as agonists at specific receptors (CMKLR1, BLT1, ALX/FPR2 and GPR32), resolvins can signal for potent counter-regulatory effects on leukocyte functions, including preventing uncontrolled neutrophil swarming, decreasing the generation of cytokines, chemokines and reactive oxygen species and promoting clearance of apoptotic neutrophils from inflamed tissues. Hence, resolvins provide mechanisms for cytoprotection of host tissues to the potentially detrimental effects of unresolved inflammation. This review highlights recent experimental findings in resolvin research, and the impact of these stereospecific molecules on the resolution of pulmonary inflammation and tissue catabasis.

1. Introduction

Acute inflammation is an essential host response to danger signals, including potential infection, noxious stimulus or tissue injury [1]. However, non-resolving inflammation is linked to many chronic inflammatory diseases, including asthma, rheumatoid arthritis, inflammatory bowel disease, and psoriasis (reviewed in [2]). In association with clinical symptoms in these chronic inflammatory diseases are elevated levels of pro-inflammatory mediators [3]. The overall magnitude and duration of inflammation depends on competing physiological processes, namely pro-phlogistic mechanisms that amplify inflammation and endogenous braking programs that control the resolution of inflammation (reviewed in [4]). In health, progression through the resolution phase of inflammation is primarily driven by the orderly phagocytic clearance of apoptotic granulocytes and debris by macrophages [5,6]. It is now established that resolution of inflammation is an active coordinated process that is spatiotemporally controlled by endogenously generated autacoids at sites of inflammation [4,7,8]. The enzymatic transformation of polyunsaturated fatty acids (PUFAs) during inflammation leads to the generation of specific endogenous mediators that act as potent agonists for resolution by exhibiting anti-inflammatory, pro-resolving, anti-fibrotic, anti-angiogenic and anti-infective actions (reviewed in [4]). The discovery that PUFAs are indispensable for health and dietary deficiencies can lead to clinical symptoms of diseases was first established in the late 1920s [9]. Such pioneering work suggested a specific relationship between the immunoregulatory role of PUFAs and the pathogenesis of major human diseases. As such, the identification of PUFA-derived mediators generated locally during the resolution of inflammation has led to a rapidly advancing understanding of the cellular and molecular mechanisms that are fundamental to resolution. Here, we review recent insights into the resolution of airway inflammation, and in particular, highlight anti-inflammatory and pro-resolving roles for resolvins in inflammation and pulmonary diseases.

2. Resolution of acute inflammation

Resolution is now appreciated to be an active process that terminates acute inflammation. At present, we know that efficient restoration of inflamed tissues to their basal state requires that inflammatory cells are effectively cleared and further neutrophil recruitment is abrogated. During this process, tissue neutrophils undergo apoptosis and are recognized and subsequently engulfed by phagocytic macrophages in a non-inflammatory manner [5,6]. Clearance of apoptotic neutrophils leads to the production of additional mediators that suppress the progression of inflammation and promote repair of damaged tissues [1012]. Dysregulation of this process leads to unresolved inflammation, which underlies the pathology of several chronic inflammatory disease processes [4,13,14]. Hence, resolution of inflammation requires a cellular flexibility in the affected tissues in order to re-establish a homeostatic state after a limited period of inflammation. This sequence of events is also referred to as ‘catabasis' - the reversion from a pathological state to one that is non-inflammatory in restoring tissue homeostasis [1,7].

2.1 Emerging cellular and molecular concepts of resolution

Inflammation resolution is a dynamic program that is partially dependent on the equilibrium of leukocyte ingress and egress at inflamed sites (reviewed in [15]). Several cellular and molecular mechanisms can limit the acute inflammatory response, including lipid mediator class switching [16]. During acute inflammation, early phase prostaglandin (PG) E2 and PGD2 can decrease neutrophil leukotriene (LT) generation and increase expression of 15-lipoxygenase (15-LOX) to switch LOX-derived AA metabolism to the biosynthesis of lipoxins [16], which are stop signals for neutrophil transmigration and activation, as well as go signals for macrophage clearance of apoptotic neutrophils. Combined, these actions actively promote resolution by clearing inflammatory cells from the previously inflamed tissue.

Distinct macrophage subsets, termed resolution-phase macrophages and fibroblastic macrophages, can play an active role in promoting the resolution of acute inflammation via phagocytic clearance of leukocytes [17,18]. Neutrophil clearance from mucosal tissues can also proceed by transmigration from the apical epithelial surface into the lumen via an epithelial CD55-mediated process [19] (reviewed in [20]). Mucosal epithelium can also express antimicrobial peptides and lipoxins can enhance host defense at mucosal surfaces by inducing epithelial bactericidal/permeability-increasing protein (BPI) [21]. Apoptotic inflammatory cells also serve a pro-resolving role by sequestering and scavenging extracellular chemokines via upregulation of cysteine-cysteine (CC) chemokine receptor 5 (CCR5) [22]. Pro-resolving mediators also display potent effects on tissue resident cells, including anti-angiogenic and anti-fibrotic actions [23,24]. These findings are relevant to chronic inflammatory diseases of the lungs as both angiogenesis and fibrosis are recognized features of airway remodeling in asthmatic individuals and pulmonary fibrosis, respectively [25]. There is now increasing evidence that defined signal transduction pathways relay pro-resolution signals that impact on a diverse range of cellular events that are fundamental to resolution [14,2630] (see Section 6). Targeting these pro-resolution pathways to control inflammation would constitute a novel therapeutic strategy for inflammatory disease.

2.2 Pro-resolution is distinct from anti-inflammation

As our understanding of the cellular and molecular mechanisms of inflammation has evolved, it has become apparent that anti-inflammation and pro-resolution are distinct physiological processes [31]. This concept is based on the initial identification of pro-resolving mediators generated at sites of inflammation that not only block granulocytic tissue entry and activation but also promote the clearance of inflammatory cells and debris from inflamed tissue (reviewed in [4]). An important functional distinction between endogenous pro-resolving mediators and pure anti-inflammatory mediators is the ability of pro-resolving mediators to accelerate the restitution of tissue homeostasis and clearance of inflammatory cells without concomitant immunosuppression. To this end, pro-resolving mediators signal for the recruitment of monocytes [32], uptake of apoptotic neutrophils by macrophages [33], increased chemokine scavenging capacity of apoptotic neutrophils (via CCR5 expression) [22], clearance of tissue neutrophils into the gastrointestinal mucosal lumen by CD55 expression [34] and enhanced mucosal host defense via expression of anti-microbial peptides by mucosal epithelial cells [21,35]. In a model and context-dependent manner, triggering of these pro-resolution pathways can stimulate the secretion of additional anti-inflammatory mediators, such as transforming growth factor (TGF)-β1 and interleukin (IL)-10 that also dampen the magnitude of the inflammatory response [36,37].

Dysregulation of constitutive neutrophil apoptosis may prevent the resolution of pulmonary inflammation and is implicated in several neutrophilic lung diseases, such as acute respiratory distress syndrome (ARDS) [38], cystic fibrosis [39], chronic obstructive pulmonary disease (COPD) [40] and severe asthma [41]. The resolution of established inflammation can be expedited in model systems by promoting apoptosis of neutrophils [28]. Human neutrophils express functionally active cyclin-dependent kinases (CDK) and inhibiting their function with selective CDK inhibitors can directly induce caspase-dependent neutrophil apoptosis and override pro-survival signals mediated by GM-CSF [28]. In addition, engulfment of apoptotic neutrophils by phagocytes leads to release of pro-resolving mediators that can act in an autocrine manner to accelerate tissue catabasis [12]. Hence, neutrophil lifespan at sites of inflammation may influence the rate of resolution in inflammatory lung disease. Recently, quantitative cellular and tissue indices of resolution were introduced to objectively assess the impact of perturbations on resolution [17,36]. For instance, the resolution interval (Ri) for neutrophils during an inflammatory response is defined by the time interval between maximal neutrophil extravasation and the point that neutrophil numbers decline to one-half of the maximum [36]. By definition, factors that shorten this interval (Ri) accelerate resolution while measures that increase Ri delay resolution. It is highly likely that several dynamic cellular processes that determine neutrophil lifespan at sites of pulmonary inflammation may influence the Ri. These may include constitutive neutrophil apoptosis as well as alternative non-apoptotic death programmes, such as oncosis/necrosis [42,43], pyroptosis [43], cytolysis [44], NETosis [45] and autophagic-like neutrophil death pathways [46].

Currently available anti-inflammatory agents [e.g. glucocorticoids (GCs) and anti-tumor necrosis factor-alpha therapy] for the treatment of chronic diseases target biochemical and cellular processes by antagonising key pro-inflammatory mediator pathways. However, their chronic use is associated with potentially serious side effects, including immunosuppression with increased susceptibility to a broad array of opportunistic bacterial, viral and fungal respiratory infections [47] and hospitalization for pneumonia results in a high COPD-related mortality rate amongst elderly patients [48]. Of relevance to pro-resolution, inhibitors of cyclooxygenases (COX) and lipoxygenases (LOX) can also interfere with the resolution process as these enzymes are also required for the endogenous formation of lipid-derived pro-resolving mediators. Hence, novel therapeutic agents that can promote anti-inflammation and endogenously exploit pro-resolution pathways to enhance host defense may have an advantageous therapeutic potential and would represent a novel concept in drug development (reviewed in [4], [49]).

3. PUFA-derived mediators regulate inflammation

PUFAs are essential for mammalian health. The ω-6 PUFA, arachidonic acid (AA; 20:4n-6) is incorporated into cellular phospholipids, and upon cell activation, specific phospholipase A2 enzymes hydrolyze the sn-2 fatty acyl bond of phospholipids to liberate AA. Once released, enzymatic metabolism of AA via COX or LOX give rise to bioactive mediators, such as prostaglandins, leukotrienes and lipoxins [50]. The ω-3 PUFAs, eicosapentaenoic acid (EPA; 20:5n–3) and docosahexaenoic acid (DHA; 22:6n–3) are enriched in neural and mucosal tissues [51,52] and in whole blood in healthy subjects - EPA accounts for ~0.5 to 2.8% of total fatty acids and DHA ~1.3 to 5.0% [5357]. At sites of inflammation, these PUFAs are available for enzymatic transformation to distinct families of pro-resolving mediators that can regulate inflammation [7,8,58] and will be the focus of Section 3.

3.1 ω-6 arachidonic acid metabolism and generation of pro-resolving mediators - Lipoxins

Lipoxins (LXs) are the lead family of specialized endogenous mediators that were first to be recognized to exhibit both anti-inflammatory and pro-resolving actions (reviewed in [4]). They are synthesized from AA via sequential catalytic actions of LOXs to generate trihydroxy-tetraene-containing eicosanoids (reviewed in [59]). LXs are produced during cell-cell interactions and are structurally and functionally distinct from pro-inflammatory eicosanoids that are principally generated during the onset of inflammation. LXs are endogenously generated at sites of pulmonary inflammation and elicit a multitude of cell-type specific responses that are relevant to asthma resolution.

Resolution of asthma exacerbations is characterized by clearance of the lung inflammatory infiltrate, restoration of epithelial barrier function and amelioration of airway hyper-responsiveness [60]. LXs block granulocyte chemotaxis, trans-endothelial and trans-epithelial migration [6166], suppress cytokine-mediated signalling in eosinophils [67], inhibit neutrophil azurophilic granule degranulation [68] and oxidative burst [26] and stimulate the non-phlogistic clearance of apoptotic neutrophils by macrophage phagocytosis [33]. In addition, LXs down-regulate the secretory potential of bronchial epithelial cells for pro-inflammatory cytokines [69]. Notably, many of these pro-resolving actions of LXA4 are shared by its endogenous 15R enantiomers, the aspirin-triggered 15-epimer-lipoxins (15-epi-LXs) [4,64,70,71]. Most recently, it has been shown that the endogenous synthesis of 15-epi-LXs can be regulated by statins during neutrophil-airway epithelial cell interactions in vitro and in bronchial mucosal inflammation in vivo [72].

In clinical asthma, several independent studies have reported that significantly lower levels of LXs are observed in plasma and the respiratory tract of severe asthmatics compared to patients with non-severe asthma [7376]. In addition, circulating levels of LXA4 correlate with lung function assessed by the measurement of forced expired volume in 1 sec (FEV1, % predicted) [73]. Of note, alveolar macrophages can generate lipoxins and LXA4 biosynthesis is also significantly decreased in alveolar macrophages from severe asthmatics compared to non-severe asthmatics and healthy subjects [77]. Further evidence for a bronchoprotective role for LXs comes from a clinical study where LXA4 administered to asthmatic subjects by the inhaled route effectively antagonised LTC4-induced bronchoconstriction [78]. Moreover, evidence for a role of LXs in resolution of allergic asthma derives from the actions of LXA4 and LXA4 stable analogs that block both airway hyper-responsiveness and allergic airway inflammation in a murine model of allergic asthma [64,71,79]. Collectively, these findings indicate that pro-resolution mechanisms are compromised in severe asthma and that LXs, when present, can serve as endogenous agonists of anti-inflammation and pro-resolution to modulate asthma pathobiology.

3.2 ω-3 eicosapentaenoic and docosahexaenoic acid metabolism and generation of pro-resolving mediators – Resolvins

An exciting area in pro-resolving mediator research began with the identification of resolvins [8,58]. These endogenous autacoids are generated from the ω-3 PUFAs EPA and DHA to produce E-series (RvE) and D-series (RvD) resolvins, respectively, and together, the resolvins are a novel class of mediators that act as natural agonists at specific receptors to promote resolution [15,8082]. Resolvins were first isolated from resolving exudates in murine dorsal air pouches after animals were treated with aspirin and EPA in vivo, and following co-incubation of human endothelial cells with neutrophils in vitro [8,58]. Resolvins are generated via interactions between aspirin acetylated COX-2 and LOX activities [58]. RvE1 biosynthetic intermediates can also be formed via a cytochrome P450-dependent process that is independent of aspirin [8,83]. There is now substantial evidence that resolvins elicit a multitude of endogenous anti-inflammatory, pro-resolving, anti-fibrotic, anti-angiogenic, anti-infective and anti-hyperalgesic actions (reviewed in Section 4). Primarily, these are achieved by signaling in a cell-type specific manner. Section 4 (vide infra) is devoted to reviewing the immunoregulatory actions of resolvins.

3.3 ω-3 docosahexaenoic acid metabolism and generation of pro-resolving mediators – Protectins and Maresins

Recently, additional classes of pro-resolution mediators derived from DHA have also been identified in resolving inflammatory exudates that display protective bioactivities, namely protectins and maresins (named for macrophage mediators in resolving inflammation) [84,85]. DHA-derived mediators are enzymatically generated via a LOX-initiated pathway [8486]. At sites of inflammation, DHA is rapidly converted by LOX activity to 17S- hydroperoxy-DHA and via an epoxide containing intermediate to protectins, such as PD1 (10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid) [86] (for detailed reviews, see refs [4,15]). DHA can be delivered during the early stages of an acute inflammatory response in albumin-rich edematous fluid [87]. It is noteworthy that the cellular components of the airway mucosa are enriched with DHA [52] and both 17S-hydroxy-DHA and PD1 are generated in human airways [88]. Of interest, PD1 levels decrease during acute exacerbations of asthma and the bronchoprotective actions of PD1 have recently been demonstrated in a murine model of allergic asthma in vivo [88]. Using ovalbumin-sensitized mice, PD1 (2 – 200 ng) given by the intravenous route to sensitized mice prior to aeroallergen challenge markedly suppressed bronchial hyperresponsiveness, mucus secretion, pulmonary eosinophil accumulation and proinflammatory cytokine and lipid mediator release. PD1 also elicited a pro-resolving effect on established pulmonary inflammation when given after allergen challenge, leading to significantly expedited resolution of allergic airway inflammation [88]. The pro-resolving actions of maresins have recently been demonstrated during the resolution of murine peritonitis in vivo [85] and have not yet been examined in the context of pulmonary inflammation. Thus, these findings are consistent with the ability of these ω-3 PUFA-derived mediators to act as natural agonists for the resolution of pulmonary inflammation (Figure 1).

Figure 1.

Figure 1

4. Resolvins: ω-3 PUFA-derived pro-resolving mediators

Two molecular series of resolvins have been identified and characterized, namely E- and D-series resolvins that are enzymatically derived from ω-3-derived-EPA and DHA, respectively. They were originally identified by physical methods during unbiased lipidomic analyses of resolving exudates [8,58]. It is now appreciated that resolvins elicit a range of cell-type specific responses that collectively block inflammation and promote resolution (reviewed in [4]). These catabatic actions in vitro and in vivo will be highlighted in this Section and are also summarized in Table 1.

Table 1.

Pro-resolving effects evoked by resolvins in vivo in experimental disease models.

Agonist of
resolution
Experimental
disease model
Species Pro-resolving/anti-inflammatory actions Cell Target/
Receptor
Ref.
EPA-derived E series

Resolvin E1
(RvE1)
Acute lung injury Mouse Attenuates acid-initiated ALI, blocks
neutrophil trafficking and pro-inflammatory
mediator release.
Airway
epithelium
leukocytes
[100]

Allergic asthma Mouse Expedites resolution of lung inflammation,
inhibits bronchial hyperresponsiveness, mucus
secretion, eosinophil recruitment.
Up-regulates IFN-γ and LXA4 release.
Airway
epithelium,
eosinophil
[79]
Prevents bronchial hyperresponsiveness,
mucus secretion, eosinophil accumulation and
Th2 cytokine mediator release.
[139]

Angiogenesis Mouse Decreases neutrophil recruitment and
suppresses pro-inflammatory gene expression.
Corneal
epithelium,
neutrophil,
macrophage /
ChemR23
[107]

Colitis Mouse Decreases neutrophil recruitment and down-
regulates pro-inflammatory gene expression.
Gut epithelium,
neutrophil,
macrophage /
ChemR23
[97,128]

Diabetes Mouse Upregulates adipokines and induces insulin
regulatory genes glucose transport (GLUT)-4
and insulin receptor substrate (IRS)-1. Protects
liver from obesity-induced insulin resistance
and hepatic steatosis.
[149]

Dorsal air pouch Mouse Cessation of neutrophil trafficking, attenuates
transepithelial/transendothelial migration.
Neutrophil /
BLT1
[8,58]

Inflammatory Pain Mouse Alleviates inflammatory pain induced by
tissue injury, nerve injury and capsaicin
treatment. Cessation of neutrophil trafficking,
reduces paw edema, blocks pro-inflammatory
mediator release.
Dorsal root
ganglion/
spinal cord
neurons /
ChemR23
[150]

Ischaemia-
reperfusion injury
in the heart
Rat Cardio-protective actions against ischaemia-
reperfusion injury and limits infarct size on
myocardium.
Cardiomyocytes [136]

Periodontitis Rabbit Cessation of neutrophil trafficking, inhibits
connective tissue and bone loss, modulates
osteoclast differentiation and bone resorption
Neutrophil,
osteoclasts /
BLT1
[113,151,
152]
Mouse

Peritonitis Mouse Cessation of neutrophil trafficking, regulates
chemokine/cytokine production. Stimulates
phagocytic clearance of apoptotic neutrophils.
Initiates phenotypic switching of macrophages
Neutrophil,
dendritic cell,
macrophage /
BLT1
ChemR23
[17,22,36,
80,83,87]

Pneumonia Mouse Promotes clearance of E. coli pneumonia,
blocks neutrophil trafficking and pro-
inflammatory mediator release, reduces
mortality.
Airway
epithelium
[100]

Retinopathy Mouse Diminishes vaso-obliteration and
neovascularisation.
Choroid-retinal
endothelial cells
[153]

Resolvin E2
(RvE2)
Peritonitis Mouse Cessation of neutrophil trafficking. Neutrophil [92]

DHA-derived D-series

Resolvin D1
(RvD1)
Angiogenesis Mouse Decreases neutrophil recruitment and counter-
regulates pro-inflammatory gene expression.
Corneal
epithelium,
neutrophil,
macrophage
[107]

Dorsal air pouch Mouse Cessation of neutrophil trafficking, attenuates
transepithelial/transendothelial migration.
Neutrophil [58,84]

Inflammatory Pain Mouse Lowers amplitude of carrageenan and
complete Freund's adjuvant-evoked heat
hyperalgesia.
Dorsal root
ganglion/
spinal cord
neurons /
ChemR23
[150]
Reduces incision-induced post-operative pain.

Ischaemia-
reperfusion injury
in the lung and
kidney
Mouse Limits neutrophil trafficking to protect against
ischaemia–reperfusion damage. Exerts anti-
fibrotic actions to regulate macrophage
function.
Neutrophil,
macrophage
[87,103]

Peritonitis Mouse Cessation of neutrophil trafficking, regulates
chemokine/cytokine production, stimulates
phagocytic clearance of apoptotic neutrophils.
Protects against oxidative stress-initiated
inflammation.
Neutrophil,
dendritic cell,
macrophage
[84,87,90,
94]

Retinopathy Mouse Diminishes vaso-obliteration and
neovascularisation.
Choroid-retinal
endothelial cells
[153]

Resolvin D2
(RvD2)
Sepsis Mouse Cessation of neutrophil trafficking by
inhibiting neutrophil–endothelial interaction,
blocks cytokine production. Stimulates
phagocytic clearance of apoptotic neutrophils.
Neutrophil,
macrophage,
endothelial cell
[94]

4.1 Lipidomic analysis of resolving exudates to identify resolvins

The use of highly sensitive and specific analytical techniques, such as liquid chromatography ultraviolet/tandem mass spectrometry (LC-UV-MS-MS), has enabled lipidomic profiling of materials from inflammatory sites. The mouse dorsal air pouch is an experimental model of a dermal wound that provides a creative system for investigation of cellular and biochemical effectors of resolution during a self-limited acute inflammatory response. Inflammatory exudates were extracted and subjected to LC-UV-MS-MS-based lipidomic analyses by Serhan and colleagues, leading to the identification of E- and D-series resolvins (reviewed in [89]). Detailed matching studies for pro-resolving actions were performed to confirm the structure-activity relationships for each endogenous compound. For elucidation of stereochemical conformation of resolvins, retrograde analyses were performed using materials generated via biogenic and total organic synthesis [8,58,83,90]. Thus, application of these sensitive and specific analytical methodologies has uncovered resolvins and additional classes of pro-resolving mediators [91].

4.2 EPA-derived E-series resolvins

Two members of the E-series resolvins have been identified and are named resolvin E1 (RvE1) and resolvin E2 (RvE2). Both are endogenously generated from EPA and were initially isolated from in vitro and in vivo models of resolution [8,58]. Within the human vasculature, transcellular synthesis of RvE1 proceeds in the presence of aspirin with transformation of C20:5 to 18R-HEPE (18R-hydroxyeicosapentaenoic acid) by aspirin-acetylated COX-2 in endothelial cells. 18R-HEPE is subsequently converted by leukocyte 5-LOX to RvE1 via a 5(6) epoxide-containing biosynthetic intermediate [8,83] (Figure 2). In humans, RvE1 levels are increased in individuals taking aspirin and/or following dietary EPA supplementation [83]. The stereochemical assignment for RvE1 is 5S,12R,18R-trihydroxyeicosa-6Z,8E,10E,14Z,16-EPA [8,83]. Functionally, RvE1’s actions are highly stereoselective both in vivo and in vitro [4]. RvE2 (5S,18(R/S)-dihydroxy-8Z,11Z,14Z,16E-eicosapentaenoic acid) is a structurally distinct member of the E-series resolvins yet displays similar bioactivities as RvE1 [92]. Administration of RvE1 and RvE2 in nanogram amounts exhibits additive cytoprotective effects [92].

Figure 2.

Figure 2

4.3 DHA-derived D-series resolvins

The two major classes of D-series resolvins are positional isomers and have been named resolvins (RvD1 to RvD4) and aspirin-triggered (AT) resolvins (AT-RvD1 to AT-RvD4) [93]. The resolvins are enzymatically generated from DHA by 15-LOX-mediated conversion to 17S-hydroperoxyDHA (17S-HpDHA) and subsequent transformation by 5-LOX to RvD1 to RvD4 via epoxide containing intermediates (Figure 3). For the AT-resolvins, DHA is initially converted by aspirin acetylated COX-2 to 17R-HpDHA that is also a substrate for 5-LOX-mediated transformation to the epimeric form of the resolvins, namely AT-RvD1 to AT-RvD4 (Figure 4). Stereochemical conformation of RvD1, RvD2 and AT-RvD1 have been assigned as 7S,8R,17S,-trihydroxy-4Z,9E,11E,13Z,15E,19Z-DHA (RvD1), 7S,16R,17S-trihydroxy-docosa-4Z,8E,10Z,12E,14E,19Z-hexaenoic acid (RvD2) and 7S,8R,17R-trihydroxy-4Z,9E,11E,13Z,15E,19Z-DHA (AT-RvD1) [90,94]. The determination of the complete stereochemistry of RvD3, RvD4, AT-RvD2, AT-RvD3 and AT-RvD4 is the subject of ongoing research.

Figure 3.

Figure 3

Figure 4.

Figure 4

4.4 Anti-inflammatory effects of resolvins on neutrophils

Neutrophils are critical to host defense, yet uncontrolled activation and/or delayed clearance of these granulocytes from inflamed sites can lead to unwanted bystander tissue damage and contribute to the pathogenesis of inflammatory disease, in particular in the lung [2,95,96]. Hence, limiting uncontrolled activation and accumulation of neutrophils at inflamed sites is an injury-limiting component of inflammatory resolution [4,5]. RvE1 inhibits neutrophil transmigration across endothelial and epithelial barriers to prevent tissue neutrophilia [8,35,80,97], down regulates expression of key neutrophil adhesion molecules (L-selectin, CD11b/CD18) [98], blocks neutrophil oxidative burst [99], stimulates non-phlogistic clearance of apoptotic neutrophils by inducing their engulfment by macrophages [80], and terminates chemokine signaling by upregulating decoy receptors (CCR5 expression) on apoptotic neutrophils during inflammatory resolution [22]. In mucosal tissues, RvE1 can enhance neutrophil clearance from the apical epithelial surface into mucosal lumen [35]. Additionally, RvE1 markedly suppresses pulmonary neutrophil accumulation in a murine model of acid-initiated acute lung injury (ALI) and aspiration pneumonia in vivo [100], adding to the already multifaceted roles of this resolvin in regulating neutrophilic inflammation (Table 1).

In addition to RvE1, RvD1 also blocks neutrophil inflammatory responses. At the single-cell level, RvD1 displays potent anti-inflammatory actions in a microfluidic chamber to inhibit IL-8-induced neutrophil chemotaxis [87]. Nanomolar concentrations of RvD1 block leukotriene B4 (LTB4)-stimulated actin polymerization and CD11b surface expression on human neutrophils [82]. RvD1 prevents transendothelial migration of human neutrophils and at nanogram doses in vivo, it potently suppresses neutrophil infiltration in murine peritonitis and ischemia reperfusion lung second organ injury [58,87,90]. RvD2 also effectively blocks tissue neutrophil accumulation. Zymosan-stimulated neutrophil infiltration in peritonitis is markedly decreased by RvD2 at doses as low as 10 pg [94]. RvD2 decreases excessive neutrophil recruitment to inflammatory loci and controls microbial sepsis after cecal ligation and puncture [94]. RvD2 can regulate neutrophil–endothelial interactions via direct modulation of adhesion molecule expression, namely decreased agonist-induced L-selectin shedding and CD18 expression on isolated human neutrophils [94]. Thus, resolvins can regulate neutrophil function to decrease extravascular granulocyte trafficking and aid the resolution response.

4.5 Pro-resolving actions of resolvins on macrophages

Macrophages can serve many pivotal functions in tissue catabasis. Efficient phagocytic removal of apoptotic granulocytes by macrophages is a key prerequisite for resolution of inflammation. Resolvins have a profound effect on the phagocytic activity of macrophages during the resolution process. Both RvD1 and RvE1 markedly increase ingestion of apoptotic neutrophils and thymocytes in isolated macrophages in vitro [80,101]. In addition, phagocytosis of zymosan A by human macrophages is greatly potentiated by RvE1 via phosphorylation-signalling pathways [102]. RvD1 also exhibits anti-inflammatory actions on a panel of macrophage-derived pro-inflammatory cytokines that are implicated in the pathogenesis of atherosclerosis [101]. Recently, it was shown that RvD1 regulates phagocytosis in a receptor-dependent manner in human macrophages [82].

In a murine model of acute kidney injury, resolvins exert cytoprotective actions as administration of D-series resolvins before the onset of ischemia effectively attenuates morphological kidney injury and blocks lipopolysaccharide-induced activation of macrophages [103]. Resolution-phase macrophages (rM) have recently been identified and shown to possess unique pro-resolving phenotype in a murine model of peritonitis in vivo [104]. These rMs closely resemble an alternative phenotype of classically activated macrophages (M1) as they share many of same cellular markers (i.e. COX-2 and inducible nitric oxide synthase), yet differ from M1s by expression of the mannose receptor Ym-1, the enzyme arginase-1 and their ability to synthesize IL-10. Using an experimental model of peritonitis, RvE1 specifically modulated phenotypic switching of macrophages from M1 macrophages to rM macrophages [17] (Table 1). Thus, macrophages are an important target cell for the agonistic actions of resolvins, and regulation of phenotypic switching of macrophages from M1 to rM can alter the dynamics of inflammatory events and consequently expedite inflammation resolution.

4.6 Cytoprotective effects of resolvins on structural cells

The airway epithelium is an important physical barrier that regulates physiological processes, including leukocyte trafficking. In airway disease, interactions between damaged epithelium and inflammatory leukocytes contribute to the pathogenesis of common conditions, such as asthma [60,105,106]. Aberrant neutrophil-mucosal epithelial interactions are a key pathological process in such disease settings [72,106] and are likely to be re-programmed during resolution of pulmonary inflammation to prevent the accumulation of activated neutrophils. In mucosal inflammation, RvE1 can bind to its cognate receptor, chemokine-like receptor 1 (CMKLR1, see section 5.1) on epithelial cells to stimulate clearance of neutrophils into the gastrointestinal tract lumen in a CD55-dependent manner [35]. Resolvins also exert potent anti-angiogenesis activities during ocular neovascularisation as topical administration of RvE1 or RvD1 to inflamed murine corneas markedly down-regulates mRNA expression levels of TNF-α, IL-1 (alpha and beta) as well as vascular endothelial growth factors (A and C) in corneal epithelial and stroma-endothelial layers [107]. Comparable results have been obtained in a co-culture model of posterior ocular inflammation where pre-treatment with RvE1 significantly inhibited the release of pro-inflammatory chemokines in choroid-retinal endothelial cells alone and following their co-culture with neutrophils and monocytes in vitro [108].

It is well established that atherosclerosis is associated with vascular inflammation with endothelial cells expressing pro-inflammatory mediators and cell adhesion molecules that promote leukocyte adherence and extravasation [109]. Recent evidence supports a protective anti-atherogenic role for resolvins. RvD1 produces significant inhibition of human aortic endothelial cell release of neutrophil chemokines, including IL-8 and monocyte chemotactic protein-1 (MCP-1) [101]. Both RvE1 and RvD1 potently block extravascular infiltration of neutrophils in vivo and modulate transmigration across monolayers of various types of endothelial cells in vitro [58,84,90]. RvD2 can trigger nitric oxide production by endothelial nitric oxide synthase (eNOS) in human umbilical vein endothelial cells (HUVEC) to modulate leukocyte trafficking [94]. Of additional interest in vascular medicine, receptors for RvE1 (CMKLR1 and BLT1) have been identified on human vascular smooth muscle cells (VSMCs) [110,111]. RvE1 blocks platelet-derived growth factor (PDGF)-stimulated chemotaxis of VSMCs isolated from human saphenous vein and decreases phosphorylation of the PDGF-Rβ [110].

The inflammation-resolving actions of resolvins are manifest in a cell- and tissue-specific manner (Table 1) (reviewed in [4]). For example, in human glioma cells [58], murine bone marrow-derived dendritic cells [112], murine primary osteoclast cultures [113] and human pancreatic islet cells, RvE1 displays potent cell-type specific actions [114]. Interestingly, retinal pigment epithelial cells and neural cells subjected to oxidative stress biosynthesize DHA-derived resolvins as a cytoprotective mechanism against oxidant injury [115,116]. Taken together, modulation of pro-inflammatory indices during both in vitro and in vivo models of human diseases are consistent with a bioprotective role for resolvins in regulating cellular responses in a cell-type specific manner to promote resolution of inflammation and injury.

5. Resolvin receptors

Details of pharmacological properties of resolvins are emerging and there are multiple receptors that transduce the potent pro-resolving actions of these compounds in a cell- and tissue-specific manner.

5.1 Receptors for E-series resolvins

RvE1 exerts its effects by acting via at least two distinct G protein-coupled receptors (GPCRs): CMKLR1 and the LTB4 receptor 1 (BLT1). Originally characterized as a receptor for the chemotactic peptide, chemerin, CMKLR1 (aka ChemR23) is a Gαi-linked receptor expressed predominantly on monocytes/macrophages and plasmacytoid dendritic cells that mediate cell migration [117119]. The binding of RvE1 to CMKLR1 was characterized using saturation binding analysis on CHO cells and it was established that human CMKLR1 exhibits high affinity for RvE1 (Kd = 11.3 ± 5.4 nM) [83]. Phylogenetically, CMKLR1 shares homology with other chemoattractant receptors, including those for LXs (denoted ALX/FPR2, vide infra) and the neutrophil chemotaxins, C5a and C3a (C5aR and C3aR, respectively) [119]. RvE1 also mediates its effects through interactions with BLT1 receptors, as determined by radioligand binding analysis [81]. In humans, BLT1 receptors are expressed primarily by neutrophils, eosinophils, monocytes/macrophages, mast cells, dendritic cells, and effector T cells [120]. Binding studies have established that RvE1 binds to the human BLT1 with relatively lower affinity than CMKLR1 with a Kd value of 45 nM [81]. RvE1 effectively competes with LTB4 for BLT1 binding to functionally antagonise LTB4-mediated responses. In addition, RvE1 can serve as a potential agonist for BLT1 signaling. Hence, the pharmacology of these receptors exhibits agonist-specific adaptation where they transduce signals specific to each mediator. The RvE2 structure-activity relationship indicates that its actions are receptor-mediated, yet the molecular identity of the RvE2 receptor has not been established.

5.2 Receptors for D-series resolvins

Specific binding sites for RvD1 were recently identified on human leukocytes with subnanomolar affinity (Kd = ~ 0.2 nM) [82]. RvD1 interacts with phagocyte GPCR-32 (GPR32) as a potent agonist to signal for pro-resolving actions. In addition, RvD1 can directly activate ALX/FPR2 with high affinity (EC50 ~1.2 pM) and is as equipotent as LXA4 (EC50 ~ 1.1 pM). The ALX/FPR2 receptor is upregulated during experimental airway inflammation and lung injury to serve as a mechanism for LX-mediated catabasis [64,69]. In addition to LXs, this receptor may also serve important roles for RvD1’s pro-resolving actions in the airway and is the subject of ongoing research. Of interest, ALX/FPR2 expression is relatively decreased in eosinophils and neutrophils in severe asthma [76], raising the possibility that this condition represents a deficit in pro-resolving signaling. ALX/FPR2-deficient mice have been prepared and display ligand specific effects on leukocyte responses and experimental inflammation [121]. The other D-series resolvins demonstrate structure-activity relationships indicative of receptor-mediated signaling pathways, but these receptors have not yet been identified.

6. Intracellular signaling mechanisms for resolvins

6.1 Nuclear factor kappa B (NF-κB)

The transcription factor NF-κB is a critical regulator of innate immune responses. A wide range of pro-inflammatory cytokines, chemokines, adhesion molecules and enzymes are regulated by the NF-κB pathway [122]. In addition, NF-κB repression has been identified as a major inducer of granulocyte apoptosis [123,124], an integral part of the resolution of inflammation and is linked to the pathogenesis of chronic inflammatory lung diseases, including asthma and chronic obstructive pulmonary disease (COPD) [122,125]. Hence, interfering with NF-κB signaling pathways may represent a feasible therapeutic strategy to manipulate apoptotic programs and consequently trigger the resolution of inflammation [14,126,127]. Several lines of evidence indicate that resolvins can modulate the NF-κB signalling pathway. For instance, RvE1 can serve as an agonist at CMKLR1 to block TNF-α-induced activation of NF-κB in a concentration-dependent manner [83]. RvE1 can also attenuate proinflammatory LTB4-mediated BLT1 signaling by decreased activation of NF-κB [81]. In primary osteoclast cultures derived from murine bone marrow, RvE1 acts via BLT1 receptors to inhibit the nuclear translocation of the p50 subunit of NF-κB to modulate osteoclast differentiation and bone resorption [113]. In the distal colons of mice subjected to dextran sulfate sodium-induced colitis, RvE1 markedly inhibits phosphorylation of NF-κB at serine residue 276 and decreases levels of proinflammatory mediators [128]. In a murine model of pneumonia, RvE1 inhibits the translocation and activation of NF-κB (p65) in lung tissue homogenates and enhances bacterial clearance [100]. Collectively, these findings point to NF-κB as an important downstream site of action for resolvins to modulate inflammatory responses and signal for resolution.

6.2 Kinases

Signal transduction pathways via the phosphatidylinositol 3-kinase (PI3-K) and the MAPK family member extracellular signal-regulated kinase (ERK) have a critical role in regulating intracellular signaling events, leading to pro-inflammatory reactions that are common in chronic inflammatory lung diseases, such as asthma and COPD [129]. PI3-Ks are a family of lipid kinases that synthesize phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3), a lipid second messenger critical for the localisation and function of the downstream serine/threonine kinase, Akt (protein kinase B) (reviewed in [130]). Intracellular signalling via the PI3-K/Akt pathway plays an important role in regulating many cellular processes, especially granulocyte migration, activation and survival [131133]. Similarly, the ERK 1/2 pathway represents a point of convergence for multiple signaling events thereby regulating important cellular functions and fate of human neutrophils [127,134,135]. RvE1-CMKLR1 interactions initiate the activation of PI3-K resulting in phosphorylation of Akt and ribosomal S6 protein via mTOR signalling [102]. RvE1 activation leads to ERK phosphorylation that is inhibited by the pan-selective PI3-K inhibitor wortmannin and a specific ERK 1/2 inhibitor PD98059 [102]. In vitro, RvE1 exerts direct protective actions on cardiomyocytes by regulating apoptotic programs in cells exposed to hypoxia or hypoxia/reoxygenation [136]. It does so by increasing phosphorylation of Akt, ERK1/2, and eNOS and attenuating the levels of key pro-apoptotic proteins, including caspase-3 and Bax. Of interest, RvE1 attenuates inflammatory pain via a glutamate ionotropic receptor mechanism in spinal dorsal horn neurons through inhibition of the ERK signaling pathway [137]. Hence, resolvins can regulate these pivotal signal transduction pathways that coordinate cellular responses ranging from anti-inflammation to anti-hyperalgesia.

7. Pro-resolving actions of resolvins in experimental models of lung disease –

E- and D-series resolvins display potent bioactivities in vivo in a wide range of experimental models of ocular, oral, dermal, gastrointestinal, renal and vascular inflammation, as well as in regulating angiogenesis, ischemia–reperfusion and hyperalgesia in vivo (Table 1). Recently, experimental models of lung disease have provided insights into cellular and molecular pro-resolving actions for resolvins in pulmonary inflammation and infection, which will be highlighted in this section.

7.1 Allergic asthma

Asthma is a chronic inflammatory disease of the conducting airways (reviewed in [60,138]). The pathogenesis of asthma is notable for excess generation of pro-inflammatory cytokines, chemokines, growth factors and eicosanoids and interactions with numerous inflammatory cell types [138]. As such, an anti-inflammatory therapeutic strategy designed to target a single pro-phlogistic mediator with selective neutralizing antibodies is unlikely to be successful, as there is a considerable degree of functional redundancy and cross-regulation amongst the intricate network of pro-inflammatory mediators and cell types within the airways. No current therapies for asthma are designed to selectively promote resolution of the pulmonary inflammation. Activation of pro-resolution signaling circuitry represents an alternate therapeutic approach for the control of asthmatic airway responses. Of interest, airway mucosal epithelial cells from individuals with asthma or cystic fibrosis have depleted stores of the ω-3-PUFA DHA compared with healthy control subjects [52]. Investigators have recently explored the potential beneficial actions of the ω-3-PUFA-derived resolvins in experimental models of airway mucosal inflammation. In a murine model of allergic asthma in vivo, RvE1 administered intravenously potently inhibits the development of allergic airway inflammation and when given during the resolution phase of inflammation, RvE1 accelerates the resolution of airway inflammation, mucus metaplasia and hyper-reactivity to methacholine. [79]. These RvE1-mediated bronchoprotective actions during resolution were related to the inhibition of Th17 effector lymphocytes and increased generation of interferon-gamma (IFN-γ) and LXA4 [79]. RvE1 and an LXA4 stable analog provided additive pro-resolving actions, yet there were important differences in their mechanism of action. For example, similar to RvE1, the LXA4 analog decreased IL-17 in bronchoalveolar lavage fluids, but distinct from RvE1, the LXA4 analog did not inhibit IL-23 production or increase IFN-γ levels. Together, these findings support the presence of independent pro-resolving signaling circuits for RvE1 and LXA4 that converge on the regulation of IL-17 to hasten catabasis in this model of allergic asthma exacerbations. RvE1 is also bioactive when given intraperitoneally before and during the sensitization and aeroallergen challenge phases. RvE1 prevents the development of bronchial hyperresponsiveness, mucus eosinophil accumulation and T-helper type 2 (Th2) cytokine mediator release [e.g. IL-13] [139] (Table 1).

7.2 Acute lung injury and bacterial pneumonia

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are important conditions characterized by an over exuberant acute inflammatory response to noxious stimuli or infection (reviewed in [140]). Patients with ALI/ARDS suffer from a loss of barrier integrity in the airways and a robust neutrophil-enriched infiltration of the lung that fills the alveoli and compromises their capacity to oxygenate the tissues, leading to excess morbidity and mortality [140]. No medical therapies have proven effective in dampening acute inflammation in ALI/ARDS. To determine the endogenous mechanisms of resolution in the lung after injury, a self-limited experimental murine model was developed for acid-initiated ALI [141]. Simulating events associated with the aspiration of gastric acid, hydrochloric acid is instilled into the airway to trigger ALI and animals are closely followed for cellular, metabolic and molecular responses. Peak inflammation and leakage permeability changes in the airway occur 12–24 h after acid instillation. When RvE1 is given intravenously just prior to acid, the amplitude of the neutrophil infiltration is dampened [100]. These actions are related to significant decrements in lung cytokines and chemokines, in particular, IL-1β, IL-6 and MCP-1. In addition to injury from sterile noxious stimuli, infection of the lung is a common etiology for ALI/ARDS and can be associated with acid injury in the form of aspiration pneumonia [140]. To determine if the anti-neutrophil actions of RvE1 were also immunosuppressive, enteric bacteria (Escherichia coli) were instilled after acid injury in the presence or absence of RvE1. After acid-initiated ALI, bacterial clearance is impaired, as this injury disrupts host defense mechanisms in the airway [100]. Of interest, RvE1 markedly enhanced bacterial clearance in the lung, decreased the levels of pro-inflammatory cytokines and chemokines and improved survival after bacterial challenge [100] (Table 1). Together, these findings of RvE1-mediated decreases in neutrophil accumulation and increased bacterial clearance support the notion that the actions of resolvins are catabatic rather than immunosuppressive.

In addition to RvE1, RvD1 displays bronchoprotective effects as this compound markedly decreased ischemia–reperfusion injury of the lung [87]. Another source of ARDS/ALI and critical illness is sepsis and RvD2 displays multi-level pro-resolving actions in experimental sepsis induced by cecal ligation and puncture [94]. Elucidation of the pro-resolving mechanisms for these and other resolvins in ALI/ARDS and pulmonary inflammation is a focus of ongoing research.

8. Resolvins as potential novel pro-resolving pharmacotherapeutics in pulmonary inflammation

8.1 Resolvin stable analogs

PUFA-derived mediators are rapidly formed and rapidly inactivated by local metabolic pathways (reviewed in [4]). These compounds thus serve as autacoids with largely autocrine and paracrine effects. The pivotal metabolic pathways for resolvins have been uncovered, informing the development of stable analogs. In addition to natural resolvins, their mimetics may serve useful to enhance the compound’s bioavailability, pharmacokinetics and pharmacodynamics. For example, 19-(p-fluorophenoxy)-RvE1 is an analog of RvE1 that resists metabolic inactivation and displays efficacious bioactivity as a pro-resolving mediator [142]. Thus, resolvins provide templates for structure-based drug design for potential use as pharmacotherapeutics in chronic inflammatory lung diseases, such as asthma [49].

8.2 Novel therapies for the treatment of asthma and chronic obstructive pulmonary disease (COPD)

Asthma and COPD are both obstructive lung diseases that have distinct pathological features in terms of their structural alterations [i.e., remodeling] and inflammatory cell profiles [143]. However, as both asthma and COPD become more severe, the patterns of pulmonary inflammation become more similar mainly due to neutrophil-dominated inflammation [143,144]. Glucocorticoids (GCs) are still the mainstay therapy for controlling airway inflammation and are potent anti-inflammatory agents, in particular for eosinophils and select lymphocyte populations [145]. However, currently available therapies, including GCs, fail to halt the progression of the airway inflammation of COPD [129], or resolve the established neutrophil-enriched airway inflammation in either severe asthma or COPD. Hence, there is an important unmet medical need for additional therapies for COPD and severe, steroid-refractory asthma that can enhance the resolution of these maladaptive airway inflammatory responses.

Given the pathological overlap of neutrophil-dominant inflammation in both COPD and severe asthma, one might predict that controlling inappropriate neutrophil accumulation and/or driving neutrophil apoptosis and their timely clearance would promote the resolution of the pulmonary inflammation. Of interest, both the pro-inflammatory mediator LTB4 and the pro-resolving mediator RvE1 can interact with BLT1 receptors. LTB4 promotes neutrophil chemotaxis and activation and has been implicated to play a pathological role in COPD with markedly increased levels in the airway secretions of patients with COPD [148]. RvE1 can antagonize LTB4 activation of BLT1 receptors that are expressed on neutrophils and select populations of effector T lymphocytes. Inhibition of LTB4-mediated activation of these inflammatory cells may provide a mechanism for RvE1 to block acute inflammation and, in combination with its CMKLR1-mediated actions on macrophages, to accelerate resolution of these chronic lung diseases.

9. Conclusions

The recent discovery of resolvins, endogenously generated from essential ω-3 PUFAs, has helped to redefine our understanding of events that terminate inflammation by uncovering their pro-resolving roles in driving inflamed tissues to a non-inflammatory state. There is now substantial evidence that these stereospecific molecules play a vital role in governing the resolution of inflammation. Resolvins are enzymatically generated during cell-cell interactions and serve as mediators of resolution circuits in vivo. Resolvins are agonists for catabasis that exert regulatory actions relevant to lung inflammation. Specifically, these endogenous autacoids have a profound effect on blunting the inflammatory response by inhibiting aberrant neutrophil trafficking and activation, stimulating phagocytosis of apoptotic cells and debris by macrophages and promoting anti-angiogenic, anti-infective and anti-fibrotic actions. These multifaceted pro-resolving actions of resolvins indicate the highly specialised handling of pathophysiological changes during established inflammation in order to expedite inflammatory resolution and restore homeostasis. The integration of resolvins into the wider array of anti-inflammatory and pro-resolving signals, including lipoxins, protectins and maresins is the subject of ongoing and future investigation.

Acknowledgements

This work was supported in part by US National Institutes of Health grants AI068084, HL68669 and P50-DE016191. The authors gratefully thank Tania A. Uddin MRPharmS and Nan Chiang PhD for their assistance with the illustrations and Anna Maria Recupero for expert assistance in preparation of the manuscript.

Abbreviations

AA

arachidonic acid

ALI

acid lung injury

ALX/FPR2

lipoxin A4 receptor/ Formyl peptide receptor 2

ARDS

acute respiratory distress syndrome

ATL

aspirin-triggered lipoxin

AT-RvD1

aspirin-triggered-resolvin D1 (7S, 8R, 17R-trihydroxy-docosa-4Z, 9E, 11E, 13Z,15E, 19Z-hexaenoic acid)

AT-RvD2

aspirin-triggered-resolvin D2 (7S, 16R, 17R-trihydroxy-docosa-4Z, 8E, 10Z,12E, 14E, 19Z-hexaenoic acid)

AT-RvD3

aspirin-triggered-resolvin D3 (4S, 11R, 17R-trihydroxy-docosa-5, 7E, 9E, 13Z,15E, 19Z-hexaenoic acid)

AT-RvD4

aspirin-triggered-resolvin D4. (4S, 5R, 17R-trihydroxy-docosa-6E, 8E, 10Z, 13Z, 15E, 19Z-hexaenoic acid)

BALF

bronchoalveolar lavage fluid

BLT1

LTB4 receptor

BPI

bactericidal/permeability-increasing protein

CDK

cyclin-dependent kinases

CHO

chinese hamster ovary

CMKLR1

chemokine receptor-like 1

COPD

chronic obstructive pulmonary disease

COX-2

cyclooxygenase-2

DHA

docosahexaenoic acid

eNOS

endothelial nitric oxide synthase

EPA

eicosapentaenoic acid

fMLP

N-formyl-methionyl-leucyl-phenylalanine

GCs

Glucocorticoids

GM-CSF

Granulocyte-macrophage colony-stimulating factor

GPCR

G protein-coupled receptor

HUVEC

human umbilical vein endothelial cells

LC-UV-MS/MS

liquid chromatography-ultraviolet spectrometry-tandem mass spectrometry

LOX

lipoxygenase

LTB4

leukotriene B4 (5S, 12R-dihydroxy-eicosa-6Z, 8E, 10E, 14Z-tetraenoic acid)

LXA4

lipoxin A4 (5S, 6R, 15S-trihydroxy-eicosa-7E, 9E, 11Z, 13E-tetraenoic acid)

M1

classically activated macrophages

mTOR

mammalian target of rapamycin

NF-κB

nuclear factor kappa B

ω-3

omega-3

ω-6

omega-6

PD1

protectin D1 (10R, 17S-dihydroxy-docosa-4Z, 7Z, 11E, 13E, 15Z, 19Z-hexaenoic acid)

PDGF

platelet-derived growth factor

PI3-K

phosphatidylinositol 3'-kinase

PUFA

polyunsaturated fatty acid

rM

resolution-phase macrophages

RvD1

resolvin D1 (7S, 8R, 17S-trihydroxy-4Z, 9E, 11E, 13Z, 15E, 19Z-docosahexaenoic acid)

RvD2

resolvin D2 (7S, 16R, 17S-trihydroxy-docosa-4Z,8E, 10Z, 12E, 14E, 19Z-hexaenoicacid)

RvD3

resolvin resolvin D3 (4S, 11, 17S-trihydroxy-5E, 7E, 9E, 13Z, 15E, 19Z-docosahexaenoic acid)

RvD4

resolvin D4 (4S, 5, 17S-trihydroxy-6E, 8E, 10Z, 13Z, 15E, 19Z-docosahexaenoic acid)

RvE1

resolvin E1 (5S,12R,18R-trihydroxy-6Z,8E,10E,14Z,16E-eicosapentaenoic acid)

RvE2

resolvin E2 (5S,18R-dihydroxy-8Z,11Z,14Z,16E-eicosapentaenoic acid

TGF-β

transforming growth factor -beta

TNF-α

tumor necrosis factor-alpha

VSMCs

vascular smooth muscle cells

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Majno GJI. Cells, Tissues and Disease: Principles of General Pathology. Blackwell: Cambridge, Mass; 1996. [Google Scholar]
  • 2.Nathan C, Ding A. Nonresolving inflammation. Cell. 140:871–882. doi: 10.1016/j.cell.2010.02.029. [DOI] [PubMed] [Google Scholar]
  • 3.Brennan FM, Maini RN, Feldmann M. Cytokine expression in chronic inflammatory disease. Br Med Bull. 1995;51:368–384. doi: 10.1093/oxfordjournals.bmb.a072967. [DOI] [PubMed] [Google Scholar]
  • 4.Serhan CN. Resolution phase of inflammation: novel endogenous anti-inflammatory and proresolving lipid mediators and pathways. Annu Rev Immunol. 2007;25:101–137. doi: 10.1146/annurev.immunol.25.022106.141647. [DOI] [PubMed] [Google Scholar]
  • 5.Savill JS, Wyllie AH, Henson JE, Walport MJ, Henson PM, Haslett C. Macrophage phagocytosis of aging neutrophils in inflammation. Programmed cell death in the neutrophil leads to its recognition by macrophages. J Clin Invest. 1989;83:865–875. doi: 10.1172/JCI113970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Savill J. A poptosisPhagocytic docking without shocking. Nature. 1998;392:442–443. doi: 10.1038/33025. [DOI] [PubMed] [Google Scholar]
  • 7.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6:1191–1197. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 8.Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang N, Gronert K. Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinflammatory drugs and transcellular processing. Journal of Experimental Medicine. 2000;192:1197–1204. doi: 10.1084/jem.192.8.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Burr GOBM. A new deficiency disease produced by the rigid exclusion of fat from the diet. J Biol Chem. 1929;82:345–367. [Google Scholar]
  • 10.Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998;101:890–898. doi: 10.1172/JCI1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature. 2000;407:784–788. doi: 10.1038/35037722. [DOI] [PubMed] [Google Scholar]
  • 12.Freire-de-Lima CG, Xiao YQ, Gardai SJ, Bratton DL, Schiemann WP, Henson PM. Apoptotic cells, through transforming growth factor-beta, coordinately induce anti-inflammatory and suppress pro-inflammatory eicosanoid and NO synthesis in murine macrophages. J Biol Chem. 2006;281:38376–38384. doi: 10.1074/jbc.M605146200. [DOI] [PubMed] [Google Scholar]
  • 13.Haslett C. Granulocyte apoptosis and its role in the resolution and control of lung inflammation. Am J Respir Crit Care Med. 1999;160:S5–S11. doi: 10.1164/ajrccm.160.supplement_1.4. [DOI] [PubMed] [Google Scholar]
  • 14.Gilroy DW, Lawrence T, Perretti M, Rossi AG. Inflammatory resolution: new opportunities for drug discovery. Nat Rev Drug Discov. 2004;3:401–416. doi: 10.1038/nrd1383. [DOI] [PubMed] [Google Scholar]
  • 15.Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nature Reviews Immunology. 2008;8:349–361. doi: 10.1038/nri2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid mediator class switching during acute inflammation: signals in resolution. Nat Immunol. 2001;2:612–619. doi: 10.1038/89759. [DOI] [PubMed] [Google Scholar]
  • 17.Navarro-Xavier RA, Newson J, Silveira VL, Farrow SN, Gilroy DW, Byström J. A new strategy for the identification of novel molecules with targeted proresolution of inflammation properties. J Immunol. 184:1516–1525. doi: 10.4049/jimmunol.0902866. [DOI] [PubMed] [Google Scholar]
  • 18.Hou TZ, Byström J, Sherlock JP, Qureshi O, Parnell SM, Anderson G, Gilroy DW, Buckley CD. A distinct subset of podoplanin (gp38) expressing F4/80+ macrophages mediate phagocytosis and are induced following zymosan peritonitis. FEBS Lett. 2010 doi: 10.1016/j.febslet.2010.07.053. [DOI] [PubMed] [Google Scholar]
  • 19.Louis NA, Hamilton KE, Kong T, Colgan SP. HIF-dependent induction of apical CD55 coordinates epithelial clearance of neutrophils. FASEB J. 2005;19:950–959. doi: 10.1096/fj.04-3251com. [DOI] [PubMed] [Google Scholar]
  • 20.Uller L, Persson CG, Erjefalt JS. Resolution of airway disease: removal of inflammatory cells through apoptosis, egression or both? Trends Pharmacol Sci. 2006;27:461–466. doi: 10.1016/j.tips.2006.07.006. [DOI] [PubMed] [Google Scholar]
  • 21.Canny G, Levy O, Furuta GT, Narravula-Alipati S, Sisson RB, Serhan CN, Colgan SP. Lipid mediator-induced expression of bactericidal/ permeability-increasing protein (BPI) in human mucosal epithelia. Proc Natl Acad Sci U S A. 2002;99:3902–3907. doi: 10.1073/pnas.052533799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ariel A, Fredman G, Sun YP, Kantarci A, Van Dyke TE, Luster AD, Serhan CN. Apoptotic neutrophils and T cells sequester chemokines during immune response resolution through modulation of CCR5 expression. Nat Immunol. 2006;7:1209–1216. doi: 10.1038/ni1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cezar-de-Mello PF, Vieira AM, Nascimento-Silva V, Villela CG, Barja-Fidalgo C, Fierro IM. ATL-1, an analogue of aspirin-triggered lipoxin A4, is a potent inhibitor of several steps in angiogenesis induced by vascular endothelial growth factor. Br J Pharmacol. 2008;153:956–965. doi: 10.1038/sj.bjp.0707650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sodin-Semrl S, Taddeo B, Tseng D, Varga J, Fiore S. Lipoxin A4 inhibits IL-1 beta-induced IL-6, IL-8, and matrix metalloproteinase-3 production in human synovial fibroblasts and enhances synthesis of tissue inhibitors of metalloproteinases. Journal of Immunology. 2000;164:2660–2666. doi: 10.4049/jimmunol.164.5.2660. [DOI] [PubMed] [Google Scholar]
  • 25.Belvisi MG, Mitchell JA. Targeting PPAR receptors in the airway for the treatment of inflammatory lung disease. Br J Pharmacol. 2009;158:994–1003. doi: 10.1111/j.1476-5381.2009.00373.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Levy BD, Fokin VV, Clark JM, Wakelam MJ, Petasis NA, Serhan CN. Polyisoprenyl phosphate (PIPP) signaling regulates phospholipase D activity: a 'stop' signaling switch for aspirin-triggered lipoxin A4. FASEB Journal. 1999;13:903–911. doi: 10.1096/fasebj.13.8.903. [DOI] [PubMed] [Google Scholar]
  • 27.Lawrence T, Bebien M, Liu GY, Nizet V, Karin M. IKKalpha limits macrophage NF-kappaB activation and contributes to the resolution of inflammation. Nature. 2005;434:1138–1143. doi: 10.1038/nature03491. [DOI] [PubMed] [Google Scholar]
  • 28.Rossi AG, Sawatzky DA, Walker A, Ward C, Sheldrake TA, Riley NA, Caldicott A, Martinez-Losa M, Walker TR, Duffin R, Gray M, Crescenzi E, Martin MC, Brady HJ, Savill JS, Dransfield I, Haslett C. Cyclin-dependent kinase inhibitors enhance the resolution of inflammation by promoting inflammatory cell apoptosis. Nat Med. 2006;12:1056–1064. doi: 10.1038/nm1468. [DOI] [PubMed] [Google Scholar]
  • 29.Leitch AE, Riley NA, Sheldrake T, Festa M, Fox S, Duffin R, Haslett C, Rossi AG. The cyclin-dependent kinase inhibitor R-roscovitine down-regulates Mcl-1 to override pro-inflammatory signalling and drive neutrophil apoptosis. Eur J Immunol. doi: 10.1002/eji.200939664. [DOI] [PubMed] [Google Scholar]
  • 30.Perretti M, Chiang N, La M, Fierro IM, Marullo S, Getting SJ, Solito E, Serhan CN. Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nature Medicine. 2002;8:1296–1302. doi: 10.1038/nm786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Serhan CN, Brain SD, Buckley CD, Gilroy DW, Haslett C, O'Neill LA, Perretti M, Rossi AG, Wallace JL. Resolution of inflammation: state of the art, definitions and terms. FASEB J. 2007;21:325–332. doi: 10.1096/fj.06-7227rev. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Maddox JF, Serhan CN. Lipoxin A4 and B4 are potent stimuli for human monocyte migration and adhesion: selective inactivation by dehydrogenation and reduction. Journal of Experimental Medicine. 1996;183:137–146. doi: 10.1084/jem.183.1.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Godson C, Mitchell S, Harvey K, Petasis NA, Hogg N, Brady HR. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. Journal of Immunology. 2000;164:1663–1667. doi: 10.4049/jimmunol.164.4.1663. [DOI] [PubMed] [Google Scholar]
  • 34.Lawrence DW, Bruyninckx WJ, Louis NA, Lublin DM, Stahl GL, Parkos CA, Colgan SP. Antiadhesive role of apical decay-accelerating factor (CD55) in human neutrophil transmigration across mucosal epithelia. J Exp Med. 2003;198:999–1010. doi: 10.1084/jem.20030380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Campbell EL, Louis NA, Tomassetti SE, Canny GO, Arita M, Serhan CN, Colgan SP. Resolvin E1 promotes mucosal surface clearance of neutrophils: a new paradigm for inflammatory resolution. FASEB J. 2007;21:3162–3170. doi: 10.1096/fj.07-8473com. [DOI] [PubMed] [Google Scholar]
  • 36.Bannenberg GL, Chiang N, Ariel A, Arita M, Tjonahen E, Gotlinger KH, Hong S, Serhan CN. Molecular circuits of resolution: formation and actions of resolvins and protectins. J Immunol. 2005;174:4345–4355. doi: 10.4049/jimmunol.174.7.4345. [DOI] [PubMed] [Google Scholar]
  • 37.Souza DG, Fagundes CT, Amaral FA, Cisalpino D, Sousa LP, Vieira AT, Pinho V, Nicoli JR, Vieira LQ, Fierro IM, Teixeira MM. The required role of endogenously produced lipoxin A4 and annexin-1 for the production of IL-10 and inflammatory hyporesponsiveness in mice. J Immunol. 2007;179:8533–8543. doi: 10.4049/jimmunol.179.12.8533. [DOI] [PubMed] [Google Scholar]
  • 38.Matute-Bello G, Liles WC, Radella F, 2nd, Steinberg KP, Ruzinski JT, Jonas M, Chi EY, Hudson LD, Martin TR. Neutrophil apoptosis in the acute respiratory distress syndrome. Am J Respir Crit Care Med. 1997;156:1969–1977. doi: 10.1164/ajrccm.156.6.96-12081. [DOI] [PubMed] [Google Scholar]
  • 39.McKeon DJ, Condliffe AM, Cowburn AS, Cadwallader KC, Farahi N, Bilton D, Chilvers ER. Prolonged survival of neutrophils from patients with Delta F508 CFTR mutations. Thorax. 2008;63:660–661. doi: 10.1136/thx.2008.096834. [DOI] [PubMed] [Google Scholar]
  • 40.Brown V, Elborn JS, Bradley J, Ennis M. Dysregulated apoptosis and NFkappaB expression in COPD subjects. Respir Res. 2009;10:24. doi: 10.1186/1465-9921-10-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Uddin M, Nong G, Ward J, Seumois G, Prince LR, Wilson SJ, Cornelius V, Dent G, Djukanovic R. Prosurvival activity for airway neutrophils in severe asthma. Thorax. 65:684–689. doi: 10.1136/thx.2009.120741. [DOI] [PubMed] [Google Scholar]
  • 42.Dacheux D, Toussaint B, Richard M, Brochier G, Croize J, Attree I. Pseudomonas aeruginosa cystic fibrosis isolates induce rapid, type III secretion-dependent, but ExoU-independent, oncosis of macrophages and polymorphonuclear neutrophils. Infect Immun. 2000;68:2916–2924. doi: 10.1128/iai.68.5.2916-2924.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fink SL, Cookson BT. Apoptosis, pyroptosis, and necrosis: mechanistic description of dead and dying eukaryotic cells. Infect Immun. 2005;73:1907–1916. doi: 10.1128/IAI.73.4.1907-1916.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bratt J, Gyllenhammar H. The role of nitric oxide in lipoxin A4-induced polymorphonuclear neutrophil-dependent cytotoxicity to human vascular endothelium in vitro. Arthritis Rheum. 1995;38:768–776. doi: 10.1002/art.1780380609. [DOI] [PubMed] [Google Scholar]
  • 45.Yousefi S, Mihalache C, Kozlowski E, Schmid I, Simon HU. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009;16:1438–1444. doi: 10.1038/cdd.2009.96. [DOI] [PubMed] [Google Scholar]
  • 46.von Gunten S, Simon HU. Autophagic-like cell death in neutrophils induced by autoantibodies. Autophagy. 2007;3:67–68. doi: 10.4161/auto.3436. [DOI] [PubMed] [Google Scholar]
  • 47.Taylor AE, Finney-Hayward TK, Quint JK, Thomas CM, Tudhope SJ, Wedzicha JA, Barnes PJ, Donnelly LE. Defective macrophage phagocytosis of bacteria in COPD. Eur Respir J. 35:1039–1047. doi: 10.1183/09031936.00036709. [DOI] [PubMed] [Google Scholar]
  • 48.Ernst P, Gonzalez AV, Brassard P, Suissa S. Inhaled Corticosteroid Use in Chronic Obstructive Pulmonary Disease and the Risk of Hospitalization for Pneumonia. Am J Respir Crit Care Med. 2007;176:162–166. doi: 10.1164/rccm.200611-1630OC. [DOI] [PubMed] [Google Scholar]
  • 49.Serhan CN, Chiang N. Endogenous pro-resolving and anti-inflammatory lipid mediators: a new pharmacologic genus. Br J Pharmacol. 2008;153 Suppl 1:S200–S215. doi: 10.1038/sj.bjp.0707489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Samuelsson B, Dahlen SE, Lindgren JA, Rouzer CA, Serhan CN. Leukotrienes and lipoxins: structures, biosynthesis, and biological effects. Science. 1987;237:1171–1176. doi: 10.1126/science.2820055. [DOI] [PubMed] [Google Scholar]
  • 51.Bazan NG. Synaptic lipid signaling: significance of polyunsaturated fatty acids and platelet-activating factor. J Lipid Res. 2003;44:2221–2233. doi: 10.1194/jlr.R300013-JLR200. [DOI] [PubMed] [Google Scholar]
  • 52.Freedman SD, Blanco PG, Zaman MM, Shea JC, Ollero M, Hopper IK, Weed DA, Gelrud A, Regan MM, Laposata M, Alvarez JG, O'Sullivan BP. Association of cystic fibrosis with abnormalities in fatty acid metabolism. N Engl J Med. 2004;350:560–569. doi: 10.1056/NEJMoa021218. [DOI] [PubMed] [Google Scholar]
  • 53.Gong J, Rosner B, Rees DG, Berson EL, Weigel-DiFranco CA, Schaefer EJ. Plasma docosahexaenoic acid levels in various genetic forms of retinitis pigmentosa. Invest Ophthalmol Vis Sci. 1992;33:2596–2602. [PubMed] [Google Scholar]
  • 54.Newcomer LM, King IB, Wicklund KG, Stanford JL. The association of fatty acids with prostate cancer risk. Prostate. 2001;47:262–268. doi: 10.1002/pros.1070. [DOI] [PubMed] [Google Scholar]
  • 55.Albert CM, Campos H, Stampfer MJ, Ridker PM, Manson JE, Willett WC, Ma J. Blood levels of long-chain n-3 fatty acids and the risk of sudden death. N Engl J Med. 2002;346:1113–1118. doi: 10.1056/NEJMoa012918. [DOI] [PubMed] [Google Scholar]
  • 56.Kew S, Mesa MD, Tricon S, Buckley R, Minihane AM, Yaqoob P. Effects of oils rich in eicosapentaenoic and docosahexaenoic acids on immune cell composition and function in healthy humans. Am J Clin Nutr. 2004;79:674–681. doi: 10.1093/ajcn/79.4.674. [DOI] [PubMed] [Google Scholar]
  • 57.Wakai K, Ito Y, Kojima M, Tokudome S, Ozasa K, Inaba Y, Yagyu K, Tamakoshi A. Intake frequency of fish and serum levels of long-chain n-3 fatty acids: a cross-sectional study within the Japan Collaborative Cohort Study. J Epidemiol. 2005;15:211–218. doi: 10.2188/jea.15.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, Moussignac RL. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. Journal of Experimental Medicine. 2002;196:1025–1037. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Serhan CN. Lipoxin biosynthesis and its impact in inflammatory and vascular events. Biochim Biophys Acta. 1994;1212:1–25. doi: 10.1016/0005-2760(94)90185-6. [DOI] [PubMed] [Google Scholar]
  • 60.Busse WW, Lemanske RF., Jr Asthma. N Engl J Med. 2001;344:350–362. doi: 10.1056/NEJM200102013440507. [DOI] [PubMed] [Google Scholar]
  • 61.Soyombo O, Spur BW, Lee TH. Effects of lipoxin A4 on chemotaxis and degranulation of human eosinophils stimulated by platelet-activating factor and N-formyl-L-methionyl-L-leucyl-L-phenylalanine. Allergy. 1994;49:230–234. doi: 10.1111/j.1398-9995.1994.tb02654.x. [DOI] [PubMed] [Google Scholar]
  • 62.Maddox JF, Colgan SP, Clish CB, Petasis NA, Fokin VV, Serhan CN. Lipoxin B4 regulates human monocyte/neutrophil adherence and motility: design of stable lipoxin B4 analogs with increased biologic activity. FASEB J. 1998;12:487–494. doi: 10.1096/fasebj.12.6.487. [DOI] [PubMed] [Google Scholar]
  • 63.Bandeira-Melo C, Bozza PT, Diaz BL, Cordeiro RS, Jose PJ, Martins MA, Serhan CN. Cutting edge: lipoxin (LX) A4 and aspirin-triggered 15-epi-LXA4 block allergen-induced eosinophil trafficking. Journal of Immunology. 2000;164:2267–2271. doi: 10.4049/jimmunol.164.5.2267. [DOI] [PubMed] [Google Scholar]
  • 64.Levy BD, De Sanctis GT, Devchand PR, Kim E, Ackerman K, Schmidt BA, Szczeklik W, Drazen JM, Serhan CN. Multi-pronged inhibition of airway hyper-responsiveness and inflammation by lipoxin A(4) Nature Medicine. 2002;8:1018–1023. doi: 10.1038/nm748. [DOI] [PubMed] [Google Scholar]
  • 65.Colgan SP, Serhan CN, Parkos CA, Delp-Archer C, Madara JL. Lipoxin A4 modulates transmigration of human neutrophils across intestinal epithelial monolayers. Journal of Clinical Investigation. 1993;92:75–82. doi: 10.1172/JCI116601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Serhan CN, Maddox JF, Petasis NA, Akritopoulou-Zanze I, Papayianni A, Brady HR, Colgan SP, Madara JL. Design of lipoxin A4 stable analogs that block transmigration and adhesion of human neutrophils. Biochemistry. 1995;34:14609–14615. doi: 10.1021/bi00044a041. [DOI] [PubMed] [Google Scholar]
  • 67.Starosta V, Pazdrak K, Boldogh I, Svider T, Kurosky A. Lipoxin A4 counterregulates GM-CSF signaling in eosinophilic granulocytes. J Immunol. 2008;181:8688–8699. doi: 10.4049/jimmunol.181.12.8688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gewirtz AT, Fokin VV, Petasis NA, Serhan CN, Madara JL. LXA4, aspirin-triggered 15-epi- LXA4, and their analogs selectively downregulate PMN azurophilic degranulation. American Journal of Physiology. 1999;276:C988–C994. doi: 10.1152/ajpcell.1999.276.4.C988. [DOI] [PubMed] [Google Scholar]
  • 69.Bonnans C, Fukunaga K, Levy MA, Levy BD. Lipoxin A(4) regulates bronchial epithelial cell responses to acid injury. American Journal of Pathology. 2006;168:1064–1072. doi: 10.2353/ajpath.2006.051056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Serhan CN. Special Issue on Lipoxins and Aspirin-Triggered Lipoxins. Prostaglandins Leukot Essent Fatty Acids. 2005;73:139–321. doi: 10.1016/j.plefa.2005.05.002. [DOI] [PubMed] [Google Scholar]
  • 71.Levy BD, Lukacs NW, Berlin AA, Schmidt B, Guilford WJ, Serhan CN, Parkinson JF. Lipoxin A4 stable analogs reduce allergic airway responses via mechanisms distinct from CysLT1 receptor antagonism. FASEB Journal. 2007;21:3877–3884. doi: 10.1096/fj.07-8653com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Planaguma A, Pfeffer MA, Rubin G, Croze R, Uddin M, Serhan CN, Levy BD. Lovastatin decreases acute mucosal inflammation via 15-epi-lipoxin A(4) Mucosal Immunol. doi: 10.1038/mi.2009.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Levy BD, Bonnans C, Silverman ES, Palmer LJ, Marigowda G, Israel E, Blood I Severe Asthma Research Program NHL. Diminished lipoxin biosynthesis in severe asthma. American Journal of Respiratory & Critical Care Medicine. 2005;172:824–830. doi: 10.1164/rccm.200410-1413OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Vachier I, Bonnans C, Chavis C, Farce M, Godard P, Bousquet J, Chanez P. Severe asthma is associated with a loss of LX4, an endogenous anti-inflammatory compound. Journal of Allergy & Clinical Immunology. 2005;115:55–60. doi: 10.1016/j.jaci.2004.09.038. [DOI] [PubMed] [Google Scholar]
  • 75.Celik GE, Erkekol FO, Misirligil Z, Melli M. Lipoxin A4 levels in asthma: relation with disease severity and aspirin sensitivity. Clinical & Experimental Allergy. 2007;37:1494–1501. doi: 10.1111/j.1365-2222.2007.02806.x. [DOI] [PubMed] [Google Scholar]
  • 76.Planaguma A, Kazani S, Marigowda G, Haworth O, Mariani TJ, Israel E, Bleecker ER, Curran-Everett D, Erzurum SC, Calhoun WJ, Castro M, Chung KF, Gaston B, Jarjour NN, Busse WW, Wenzel SE, Levy BD. Airway lipoxin A4 generation and lipoxin A4 receptor expression are decreased in severe asthma. Am J Respir Crit Care Med. 2008;178:574–582. doi: 10.1164/rccm.200801-061OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bhavsar P, Levy B, Hew M, Pfeffer M, Kazani S, Israel E, Chung K.Corticosteroid suppression of lipoxin A4 and leukotriene B4from alveolar macrophages in severe asthma Respiratory Research; 1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Christie PE, Spur BW, Lee TH. The effects of lipoxin A4 on airway responses in asthmatic subjects. American Review of Respiratory Disease. 1992;145:1281–1284. doi: 10.1164/ajrccm/145.6.1281. [DOI] [PubMed] [Google Scholar]
  • 79.Haworth O, Cernadas M, Yang R, Serhan CN, Levy BD. Resolvin E1 regulates interleukin 23, interferon-gamma and lipoxin A4 to promote the resolution of allergic airway inflammation. Nat Immunol. 2008;9:873–879. doi: 10.1038/ni.1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature. 2007;447:869–874. doi: 10.1038/nature05877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Arita M, Ohira T, Sun YP, Elangovan S, Chiang N, Serhan CN. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. Journal of Immunology. 2007;178:3912–3917. doi: 10.4049/jimmunol.178.6.3912. [DOI] [PubMed] [Google Scholar]
  • 82.Krishnamoorthy S, Recchiuti A, Chiang N, Yacoubian S, Lee CH, Yang R, Petasis NA, Serhan CN. Resolvin D1 binds human phagocytes with evidence for proresolving receptors. Proc Natl Acad Sci U S A. 107:1660–1665. doi: 10.1073/pnas.0907342107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Arita M, Bianchini F, Aliberti J, Sher A, Chiang N, Hong S, Yang R, Petasis NA, Serhan CN. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. Journal of Experimental Medicine. 2005;201:713–722. doi: 10.1084/jem.20042031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hong S, Gronert K, Devchand PR, Moussignac RL, Serhan CN. Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation. Journal of Biological Chemistry. 2003;278:14677–14687. doi: 10.1074/jbc.M300218200. [DOI] [PubMed] [Google Scholar]
  • 85.Serhan CN, Yang R, Martinod K, Kasuga K, Pillai PS, Porter TF, Oh SF, Spite M. Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions. J Exp Med. 2009;206:15–23. doi: 10.1084/jem.20081880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Serhan CN, Gotlinger K, Hong S, Lu Y, Siegelman J, Baer T, Yang R, Colgan SP, Petasis NA. Anti-inflammatory actions of neuroprotectin D1/protectin D1 and its natural stereoisomers: assignments of dihydroxy-containing docosatrienes. J Immunol. 2006;176:1848–1859. doi: 10.4049/jimmunol.176.3.1848. [DOI] [PubMed] [Google Scholar]
  • 87.Kasuga K, Yang R, Porter TF, Agrawal N, Petasis NA, Irimia D, Toner M, Serhan CN. Rapid appearance of resolvin precursors in inflammatory exudates: novel mechanisms in resolution. J Immunol. 2008;181:8677–8687. doi: 10.4049/jimmunol.181.12.8677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Levy BD, Kohli P, Gotlinger K, Haworth O, Hong S, Kazani S, Israel E, Haley KJ, Serhan CN. Protectin D1 is generated in asthma and dampens airway inflammation and hyperresponsiveness. Journal of Immunology. 2007;178:496–502. doi: 10.4049/jimmunol.178.1.496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Serhan CN, Hong S, Lu Y. Lipid mediator informatics-lipidomics: novel pathways in mapping resolution. AAPS J. 2006;8:E284–E297. doi: 10.1007/BF02854899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sun YP, Oh SF, Uddin J, Yang R, Gotlinger K, Campbell E, Colgan SP, Petasis NA, Serhan CN. Resolvin D1 and its aspirin-triggered 17R epimer. Stereochemical assignments, antiinflammatory properties, and enzymatic inactivation. Journal of Biological Chemistry. 2007;282:9323–9334. doi: 10.1074/jbc.M609212200. [DOI] [PubMed] [Google Scholar]
  • 91.Lu Y, Hong S, Tjonahen E, Serhan CN. Mediator-lipidomics: databases and search algorithms for PUFA-derived mediators. J Lipid Res. 2005;46:790–802. doi: 10.1194/jlr.D400020-JLR200. [DOI] [PubMed] [Google Scholar]
  • 92.Tjonahen E, Oh SF, Siegelman J, Elangovan S, Percarpio KB, Hong S, Arita M, Serhan CN. Resolvin E2: identification and anti-inflammatory actions: pivotal role of human 5- lipoxygenase in resolvin E series biosynthesis. Chemistry & Biology. 2006;13:1193–1202. doi: 10.1016/j.chembiol.2006.09.011. [DOI] [PubMed] [Google Scholar]
  • 93.Serhan CN, Arita M, Hong S, Gotlinger K. Resolvins, docosatrienes, and neuroprotectins, novel omega-3-derived mediators, and their endogenous aspirin-triggered epimers. Lipids. 2004;39:1125–1132. doi: 10.1007/s11745-004-1339-7. [DOI] [PubMed] [Google Scholar]
  • 94.Spite M, Norling LV, Summers L, Yang R, Cooper D, Petasis NA, Flower RJ, Perretti M, Serhan CN. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis. Nature. 2009;461:1287–1291. doi: 10.1038/nature08541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Weiss SJ. Tissue destruction by neutrophils. N Engl J Med. 1989;320:365–376. doi: 10.1056/NEJM198902093200606. [DOI] [PubMed] [Google Scholar]
  • 96.Grigg JM, Savill JS, Sarraf C, Haslett C, Silverman M. Neutrophil apoptosis and clearance from neonatal lungs. Lancet. 1991;338:720–722. doi: 10.1016/0140-6736(91)91443-x. [DOI] [PubMed] [Google Scholar]
  • 97.Arita M, Yoshida M, Hong S, Tjonahen E, Glickman JN, Petasis NA, Blumberg RS, Serhan CN. Resolvin E1, an endogenous lipid mediator derived from omega-3 eicosapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:7671–7676. doi: 10.1073/pnas.0409271102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Dona M, Fredman G, Schwab JM, Chiang N, Arita M, Goodarzi A, Cheng G, von Andrian UH, Serhan CN. Resolvin E1, an EPA-derived mediator in whole blood, selectively counterregulates leukocytes and platelets. Blood. 2008;112:848–855. doi: 10.1182/blood-2007-11-122598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gronert K, Kantarci A, Levy BD, Clish CB, Odparlik S, Hasturk H, Badwey JA, Colgan SP, Van Dyke TE, Serhan CN. A molecular defect in intracellular lipid signaling in human neutrophils in localized aggressive periodontal tissue damage. J Immunol. 2004;172:1856–1861. doi: 10.4049/jimmunol.172.3.1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Seki H, Fukunaga K, Arita M, Arai H, Nakanishi H, Taguchi R, Miyasho T, Takamiya R, Asano K, Ishizaka A, Takeda J, Levy BD. The anti-inflammatory and proresolving mediator resolvin E1 protects mice from bacterial pneumonia and acute lung injury. J Immunol. 184:836–843. doi: 10.4049/jimmunol.0901809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Merched AJ, Ko K, Gotlinger KH, Serhan CN, Chan L. Atherosclerosis: evidence for impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J. 2008;22:3595–3606. doi: 10.1096/fj.08-112201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ohira T, Arita M, Omori K, Recchiuti A, Van Dyke TE, Serhan CN. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J Biol Chem. 285:3451–3461. doi: 10.1074/jbc.M109.044131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Duffield JS, Hong S, Vaidya VS, Lu Y, Fredman G, Serhan CN, Bonventre JV. Resolvin D series and protectin D1 mitigate acute kidney injury. Journal of Immunology. 2006;177:5902–5911. doi: 10.4049/jimmunol.177.9.5902. [DOI] [PubMed] [Google Scholar]
  • 104.Byström J, Evans I, Newson J, Stables M, Toor I, van Rooijen N, Crawford M, Colville-Nash P, Farrow S, Gilroy DW. Resolution-phase macrophages possess a unique inflammatory phenotype that is controlled by cAMP. Blood. 2008;112:4117–4127. doi: 10.1182/blood-2007-12-129767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Djukanovic R, Roche WR, Wilson JW, Beasley CR, Twentyman OP, Howarth RH, Holgate ST. Mucosal inflammation in asthma. Am Rev Respir Dis. 1990;142:434–457. doi: 10.1164/ajrccm/142.2.434. [DOI] [PubMed] [Google Scholar]
  • 106.Uddin M, Seumois G, Lau LC, Rytila P, Davies DE, Djukanovic R. Enhancement of neutrophil function by the bronchial epithelium stimulated by epidermal growth factor. Eur Respir J. 2008;31:714–724. doi: 10.1183/09031936.00144307. [DOI] [PubMed] [Google Scholar]
  • 107.Jin Y, Arita M, Zhang Q, Saban DR, Chauhan SK, Chiang N, Serhan CN, Dana R. Anti-angiogenesis effect of the novel anti-inflammatory and pro-resolving lipid mediators. Invest Ophthalmol Vis Sci. 2009;50:4743–4752. doi: 10.1167/iovs.08-2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Tian H, Lu Y, Sherwood AM, Hongqian D, Hong S. Resolvins E1 and D1 in choroid-retinal endothelial cells and leukocytes: biosynthesis and mechanisms of anti-inflammatory actions. Invest Ophthalmol Vis Sci. 2009;50:3613–3620. doi: 10.1167/iovs.08-3146. [DOI] [PubMed] [Google Scholar]
  • 109.Libby P, Aikawa M. Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med. 2002;8:1257–1262. doi: 10.1038/nm1102-1257. [DOI] [PubMed] [Google Scholar]
  • 110.Ho KJ, Spite M, Owens CD, Lancero H, Kroemer AH, Pande R, Creager MA, Serhan CN, Conte MS. Aspirin-Triggered Lipoxin and Resolvin E1 Modulate Vascular Smooth Muscle Phenotype and Correlate with Peripheral Atherosclerosis. Am J Pathol. doi: 10.2353/ajpath.2010.091082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Back M, Bu DX, Branström R, Sheikine Y, Yan ZQ, Hansson GK. Leukotriene B4 signaling through NF-kappaB-dependent BLT1 receptors on vascular smooth muscle cells in atherosclerosis and intimal hyperplasia. Proc Natl Acad Sci U S A. 2005;102:17501–17506. doi: 10.1073/pnas.0505845102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Vassiliou EK, Kesler OM, Tadros JH, Ganea D. Bone marrow-derived dendritic cells generated in the presence of resolvin E1 induce apoptosis of activated CD4+ T cells. J Immunol. 2008;181:4534–4544. doi: 10.4049/jimmunol.181.7.4534. [DOI] [PubMed] [Google Scholar]
  • 113.Herrera BS, Ohira T, Gao L, Omori K, Yang R, Zhu M, Muscara MN, Serhan CN, Van Dyke TE, Gyurko R. An endogenous regulator of inflammation, resolvin E1, modulates osteoclast differentiation and bone resorption. Br J Pharmacol. 2008;155:1214–1223. doi: 10.1038/bjp.2008.367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lund T, Mangsbo SM, Scholz H, Gjorstrup P, Totterman TH, Korsgren O, Foss A. Resolvin E1 Reduces Proinflammatory Markers in Human Pancreatic Islets in vitro. Exp Clin Endocrinol Diabetes. doi: 10.1055/s-0029-1241825. [DOI] [PubMed] [Google Scholar]
  • 115.Bazan NG. Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr Opin Clin Nutr Metab Care. 2007;10:136–141. doi: 10.1097/MCO.0b013e32802b7030. [DOI] [PubMed] [Google Scholar]
  • 116.Marcheselli VL, Mukherjee PK, Arita M, Hong S, Antony R, Sheets K, Winkler JW, Petasis NA, Serhan CN, Bazan NG. Neuroprotectin D1/protectin D1 stereoselective and specific binding with human retinal pigment epithelial cells and neutrophils. Prostaglandins Leukot Essent Fatty Acids. 82:27–34. doi: 10.1016/j.plefa.2009.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Samson M, Edinger AL, Stordeur P, Rucker J, Verhasselt V, Sharron M, Govaerts C, Mollereau C, Vassart G, Doms RW, Parmentier M. ChemR23, a putative chemoattractant receptor, is expressed in monocyte-derived dendritic cells and macrophages and is a coreceptor for SIV and some primary HIV-1 strains. Eur J Immunol. 1998;28:1689–1700. doi: 10.1002/(SICI)1521-4141(199805)28:05<1689::AID-IMMU1689>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
  • 118.Wittamer V, Franssen JD, Vulcano M, Mirjolet JF, Le Poul E, Migeotte I, Brezillon S, Tyldesley R, Blanpain C, Detheux M, Mantovani A, Sozzani S, Vassart G, Parmentier M, Communi D. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J Exp Med. 2003;198:977–985. doi: 10.1084/jem.20030382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Cash JL, Hart R, Russ A, Dixon JP, Colledge WH, Doran J, Hendrick AG, Carlton MB, Greaves DR. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J Exp Med. 2008;205:767–775. doi: 10.1084/jem.20071601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yokomizo T, Izumi T, Chang K, Takuwa Y, Shimizu T. A G-protein-coupled receptor for leukotriene B4 that mediates chemotaxis. Nature. 1997;387:620–624. doi: 10.1038/42506. [DOI] [PubMed] [Google Scholar]
  • 121.Dufton N, Hannon R, Brancaleone V, Dalli J, Patel HB, Gray M, D’Acquisto F, Buckingham JC, Perretti M, Flower RJ. Anti-inflammatory role of the murine formyl-Peptide receptor 2: ligand-specific effects on leukocyte responses and experimental inflammation. J Immunol. 184:2611–2619. doi: 10.4049/jimmunol.0903526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med. 1997;336:1066–1071. doi: 10.1056/NEJM199704103361506. [DOI] [PubMed] [Google Scholar]
  • 123.Ward C, Chilvers ER, Lawson MF, Pryde JG, Fujihara S, Farrow SN, Haslett C, Rossi AG. NF-kappaB activation is a critical regulator of human granulocyte apoptosis in vitro. J Biol Chem. 1999;274:4309–4318. doi: 10.1074/jbc.274.7.4309. [DOI] [PubMed] [Google Scholar]
  • 124.Fujihara S, Ward C, Dransfield I, Hay RT, Uings IJ, Hayes B, Farrow SN, Haslett C, Rossi AG. Inhibition of nuclear factor-kappaB activation un-masks the ability of TNF-alpha to induce human eosinophil apoptosis. Eur J Immunol. 2002;32:457–466. doi: 10.1002/1521-4141(200202)32:2<457::AID-IMMU457>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 125.Hart LA, Krishnan VL, Adcock IM, Barnes PJ, Chung KF. Activation and localization of transcription factor, nuclear factor-kappaB, in asthma. Am J Respir Crit Care Med. 1998;158:1585–1592. doi: 10.1164/ajrccm.158.5.9706116. [DOI] [PubMed] [Google Scholar]
  • 126.Rossi AG, Hallett JM, Sawatzky DA, Teixeira MM, Haslett C. Modulation of granulocyte apoptosis can influence the resolution of inflammation. Biochem Soc Trans. 2007;35:288–291. doi: 10.1042/BST0350288. [DOI] [PubMed] [Google Scholar]
  • 127.Sawatzky DA, Willoughby DA, Colville-Nash PR, Rossi AG. The involvement of the apoptosis-modulating proteins ERK 1/2, Bcl-xL and Bax in the resolution of acute inflammation in vivo. Am J Pathol. 2006;168:33–41. doi: 10.2353/ajpath.2006.050058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ishida T, Yoshida M, Arita M, Nishitani Y, Nishiumi S, Masuda A, Mizuno S, Takagawa T, Morita Y, Kutsumi H, Inokuchi H, Serhan CN, Blumberg RS, Azuma T. Resolvin E1, an endogenous lipid mediator derived from eicosapentaenoic acid, prevents dextran sulfate sodium-induced colitis. Inflamm Bowel Dis. 16:87–95. doi: 10.1002/ibd.21029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Barnes PJ. Frontrunners in novel pharmacotherapy of COPD. Curr Opin Pharmacol. 2008;8:300–307. doi: 10.1016/j.coph.2008.03.001. [DOI] [PubMed] [Google Scholar]
  • 130.Vanhaesebroeck B, Leevers SJ, Ahmadi K, Timms J, Katso R, Driscoll PC, Woscholski R, Parker PJ, Waterfield MD. Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem. 2001;70:535–602. doi: 10.1146/annurev.biochem.70.1.535. [DOI] [PubMed] [Google Scholar]
  • 131.Bonnans C, Fukunaga K, Keledjian R, Petasis NA, Levy BD. Regulation of phosphatidylinositol 3-kinase by polyisoprenyl phosphates in neutrophil-mediated tissue injury. Journal of Experimental Medicine. 2006;203:857–863. doi: 10.1084/jem.20052143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Cantley LC. The phosphoinositide 3-kinase pathway. Science. 2002;296:1655–1657. doi: 10.1126/science.296.5573.1655. [DOI] [PubMed] [Google Scholar]
  • 133.Hawkins PT, Anderson KE, Davidson K, Stephens LR. Signalling through Class I PI3Ks in mammalian cells. Biochem Soc Trans. 2006;34:647–662. doi: 10.1042/BST0340647. [DOI] [PubMed] [Google Scholar]
  • 134.Downey GP, Butler JR, Tapper H, Fialkow L, Saltiel AR, Rubin BB, Grinstein S. Importance of MEK in neutrophil microbicidal responsiveness. J Immunol. 1998;160:434–443. [PubMed] [Google Scholar]
  • 135.Nolan B, Duffy A, Paquin L, De M, Collette H, Graziano CM, Bankey P. Mitogen-activated protein kinases signal inhibition of apoptosis in lipopolysaccharide-stimulated neutrophils. Surgery. 1999;126:406–412. [PubMed] [Google Scholar]
  • 136.Keyes KT, Ye Y, Lin Y, Zhang C, Perez-Polo JR, Gjorstrup P, Birnbaum Y. Resolvin E1 protects the rat heart against reperfusion injury. Am J Physiol Heart Circ Physiol. 299:H153–H164. doi: 10.1152/ajpheart.01057.2009. [DOI] [PubMed] [Google Scholar]
  • 137.Xu ZZ, Zhang L, Liu T, Park JY, Berta T, Yang R, Serhan CN, Ji RR. Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions. Nat Med. doi: 10.1038/nm.2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Holgate ST, Polosa R. Treatment strategies for allergy and asthma. Nat Rev Immunol. 2008;8:218–230. doi: 10.1038/nri2262. [DOI] [PubMed] [Google Scholar]
  • 139.Aoki H, Hisada T, Ishizuka T, Utsugi M, Kawata T, Shimizu Y, Okajima F, Dobashi K, Mori M. Resolvin E1 dampens airway inflammation and hyperresponsiveness in a murine model of asthma. Biochemical & Biophysical Research Communications. 2008;367:509–515. doi: 10.1016/j.bbrc.2008.01.012. [DOI] [PubMed] [Google Scholar]
  • 140.Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med. 2000;342:1334–1349. doi: 10.1056/NEJM200005043421806. [DOI] [PubMed] [Google Scholar]
  • 141.Fukunaga K, Arita M, Takahashi M, Morris AJ, Pfeffer M, Levy BD. Identification and functional characterization of a presqualene diphosphate phosphatase. Journal of Biological Chemistry. 2006;281:9490–9497. doi: 10.1074/jbc.M512970200. [DOI] [PubMed] [Google Scholar]
  • 142.Arita M, Oh SF, Chonan T, Hong S, Elangovan S, Sun YP, Uddin J, Petasis NA, Serhan CN. Metabolic inactivation of resolvin E1 and stabilization of its anti-inflammatory actions. J Biol Chem. 2006;281:22847–22854. doi: 10.1074/jbc.M603766200. [DOI] [PubMed] [Google Scholar]
  • 143.Jeffery PK. Remodeling and inflammation of bronchi in asthma and chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2004;1:176–183. doi: 10.1513/pats.200402-009MS. [DOI] [PubMed] [Google Scholar]
  • 144.Fahy JV, Kim KW, Liu J, Boushey HA. Prominent neutrophilic inflammation in sputum from subjects with asthma exacerbation. J Allergy Clin Immunol. 1995;95:843–852. doi: 10.1016/s0091-6749(95)70128-1. [DOI] [PubMed] [Google Scholar]
  • 145.Belvisi MG. Regulation of inflammatory cell function by corticosteroids. Proc Am Thorac Soc. 2004;1:207–214. doi: 10.1513/pats.200402-002MS. [DOI] [PubMed] [Google Scholar]
  • 146.Mecklenburgh K, Murray J, Brazil T, Ward C, Rossi AG, Chilvers ER. Role of neutrophil apoptosis in the resolution of pulmonary inflammation. Monaldi Arch Chest Dis. 1999;54:345–349. [PubMed] [Google Scholar]
  • 147.Savill J, Dransfield I, Gregory C, Haslett C. A blast from the past: clearance of apoptotic cells regulates immune responses. Nat Rev Immunol. 2002;2:965–975. doi: 10.1038/nri957. [DOI] [PubMed] [Google Scholar]
  • 148.Hill AT, Bayley D, Stockley RA. The interrelationship of sputum inflammatory markers in patients with chronic bronchitis. Am J Respir Crit Care Med. 1999;160:893–898. doi: 10.1164/ajrccm.160.3.9901091. [DOI] [PubMed] [Google Scholar]
  • 149.Gonzalez-Periz A, Horrillo R, Ferre N, Gronert K, Dong B, Moran-Salvador E, Titos E, Martinez-Clemente M, Lopez-Parra M, Arroyo V, Claria J. Obesity-induced insulin resistance and hepatic steatosis are alleviated by omega-3 fatty acids: a role for resolvins and protectins. FASEB J. 2009;23:1946–1957. doi: 10.1096/fj.08-125674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Xu ZZ, Zhang L, Liu T, Park JY, Berta T, Yang R, Serhan CN, Ji RR. Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions. Nat Med. 16:592–597. doi: 10.1038/nm.2123. 591p following 597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hasturk H, Kantarci A, Ohira T, Arita M, Ebrahimi N, Chiang N, Petasis NA, Levy BD, Serhan CN, Van Dyke TE. RvE1 protects from local inflammation and osteoclast- mediated bone destruction in periodontitis. FASEB J. 2006;20:401–403. doi: 10.1096/fj.05-4724fje. [DOI] [PubMed] [Google Scholar]
  • 152.Hasturk H, Kantarci A, Goguet-Surmenian E, Blackwood A, Andry C, Serhan CN, Van Dyke TE. Resolvin E1 regulates inflammation at the cellular and tissue level and restores tissue homeostasis in vivo. J Immunol. 2007;179:7021–7029. doi: 10.4049/jimmunol.179.10.7021. [DOI] [PubMed] [Google Scholar]
  • 153.Connor KM, SanGiovanni JP, Lofqvist C, Aderman CM, Chen J, Higuchi A, Hong S, Pravda EA, Majchrzak S, Carper D, Hellstrom A, Kang JX, Chew EY, Salem N, Jr, Serhan CN, Smith LE. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med. 2007;13:868–873. doi: 10.1038/nm1591. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES