Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 1.
Published in final edited form as: Clin Chest Med. 2012 Jul 25;33(3):559–570. doi: 10.1016/j.ccm.2012.06.006

Resolution of Inflammation in Asthma

Bruce D Levy a,b,*, Isabelle Vachier c, Charles Serhan d,e
PMCID: PMC3431599  NIHMSID: NIHMS397267  PMID: 22929102

Synopsis

The resolution of inflammation in healthy airways is an active process with specialized mediators and cellular mechanisms that are enlisted to restore tissue homeostasis. In this article, we will focus on recent discoveries of natural mediators derived from essential fatty acids, including omega-3 fatty acids, that have anti-inflammatory and pro-resolving actions. These specialized pro-resolving mediators serve as agonists at specific receptors. Asthma is a disease of chronic, non-resolving inflammation of the airway without known cure. While the biosynthesis of pro-resolving mediators occurs during asthma, defects in their production are associated with disease severity, suggesting that the pathobiology of asthma may result from impaired resolution of airway inflammation.

Keywords: inflammation, resolution, lipoxins, mediators, asthma

Introduction

Asthma is characterized by increased and chronic airway inflammation39. Airway sampling by bronchoscopy or sputum analysis most often reveals abundant eosinophils and activated T cells and lung histology demonstrates airway remodeling with disordered mucosal epithelium21,24. While noxious stimuli, including potential allergens, can initiate an acute inflammatory response that is self-limited, non-resolving inflammation is linked to asthma and other chronic inflammatory diseases (reviewed in 73). 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 92). In health, the resolution of inflammation is an active coordinated process that is spatiotemporally controlled by endogenously generated autacoids at sites of inflammation62. While several classes of mediators participate in resolution, the enzymatic transformations of polyunsaturated fatty acids (PUFAs) to specific pro-resolving agonists are of particular interest. The discovery that PUFAs are essential dietary constituents 23 led to the recognition of their immunoregulatory actions91. These PUFA-derived mediators display cell-type selective anti-inflammatory, pro-resolving, anti-fibrotic, anti-angiogenic and anti-infective actions (reviewed in 92).

Here, we review recent findings on new mechanisms and mediators for resolution of airway inflammation with a focus on their relevance to asthma.

Resolution of acute inflammation

The acute inflammatory response to inhaled pathogens, particles and toxins is inherently protective and essential to ultimately restoring the injured airway to its normal physiological functioning. Acute inflammation is initiated within minutes of recognition of a danger signal, and is generally self-limited, resolving within hours or days. In several conditions that are clinically recognized as lung disease, including asthma, chronic inflammation persists. Somehow, a naturally protective response becomes unrestrained and leads to chronic changes to airway structure and function. In most individuals with asthma, it is this chronic inflammation that causes the clinical expression of symptoms, namely cough, mucus, chest tightness, wheeze and shortness of breath. There is a rapidly expanding exploration of the natural mechanisms and mediators of resolution that limit acute inflammation in healthy tissues and this information has turned conventional thinking on its head regarding the pathophysiology of asthma and other chronic inflammatory diseases.

Resolution is now appreciated to be an active process that terminates acute inflammation 62,95,101. 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 85,87. Clearance of apoptotic neutrophils also leads to the production of additional mediators that suppress the progression of inflammation and promote repair of damaged tissues 38,42,86. Dysregulation of this process leads to unresolved inflammation, which underlies the pathology of several chronic inflammatory disease processes45,92. Hence, resolution of inflammation requires cellular interactions 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 to a non-inflammatory state of tissue homeostasis12,103.

In health, the acute inflammatory response is self-limited. Early tissue edema (minutes-hours) and neutrophil accumulation (hours to days) will decrease with time and lymphocytes, macrophages and other monocytoid cells will traffic to the inflamed tissue to restore homeostasis68. While anti-leukocyte actions are commonly considered anti-inflammatory, it is important to view each cell types’ roles in this dynamic process of inflammation resolution (aka catabasis). Inhibition of neutrophil transmigration and activation is anti-inflammatory while restitution of barrier integrity (endothelial, epithelial or both), recruitment of monocytoid cells, and promotion of macrophage clearance of apoptotic cells, microbes and tissue debris are all pro-resolving. In the lung, restitution of barrier integrity is of heightened importance secondary to concern for alveolar edema leading to hypoxemia. Thus, lung specific pro-resolving events include clearance of edema and transitional matrix, repopulation of the airway epithelia and restoration of pulmonary surfactants.

For resolution of adaptive immune responses, allergen-specific or pathogen-specific effector T cells and inflammatory macrophages need to be cleared from the lung. Direct and indirect mechanisms for T cell clearance include Natural Killer (NK) cell direct cytotoxicity, macrophage engulfment of apoptotic T cells and decrements in pro-inflammatory mediators.

The majority of individuals with asthma have chronic inflammation of the airways that does not resolve. Perhaps as many as 50% of adult asthmatics and an even higher proportion of pediatric asthmatic individuals have an expansion of allergy airway inflammation that is characterized by an abundance of CD4+ TH2 cells with increased interleukin (IL)-5 and IL-13 production116,117. IL-5 is a potent chemoattractant and activator of eosinophils, which are also present in large numbers in most asthmatic subjects39. Eosinophils and mast cells release significant amounts of the cysteinyl leukotrienes (CysLTs; LTC4, LTD4 and LTE4) and LTD4 is the most potent constrictor of airway smooth muscle identified to date82. These mediators also induce vascular permeability changes and tissue edema. IL-13 leads to mucous metaplasia of the bronchial epithelium with increased airway mucus114,121. Together, the increased numbers of airway leukocytes (T cells, eosinophils), pro-inflammatory (IL-5, IL-13) and pro-phlogistic (CysLTs) mediators, smooth muscle constriction and mucus production by epithelial cells lead to airway narrowing and hyperactivity.

While chronic eosinophilic bronchitis is common in asthma, the condition is syndromic and airway pathobiology is more complex in many individuals with asthma. Some experience a predominately neutrophilic infiltration, and others are pauci-immune, likely reflecting a primary disorder of airway smooth muscle72. Environmental factors, such as cigarette smoke, microbial pathogens and air pollution, can lead to mixed and more complex airway inflammation39. Despite the presence of airway inflammation in the vast majority of individuals with asthma, there is still limited understanding of why the asthmatic airway inflammation fails to resolve. In addition, potent anti-inflammatory agents such as glucocorticoids are available, yet their use does not cure asthma. Once adults are diagnosed with asthma, this is most often a life-long illness. Thus, important questions remain to be answered in determining why asthmatic inflammation fails to resolve, so that pro-resolving therapeutics can be developed.

Anti-inflammation and pro-resolution are distinct physiological processes 93. The identification of pro-resolving mediators that facilitate the clearance of inflammatory cells and debris from inflamed tissue has improved our understanding of the distinction between pro-resolving and purely anti-inflammatory mediators (reviewed in 92). While anti-inflammatory mediators block granulocyte tissue entry and activation, endogenous pro-resolving mediators accelerate the restitution of tissue homeostasis and clearance of inflammatory cells without concomitant immunosuppression. To this end, pro-resolving mediators decrease allergic edema11, recruit monocytes in a non-phlogistic fashion 67, increase phagocytosis and efferocytosis by macrophages46, increase chemokine scavenging by apoptotic neutrophils (via CCR5 expression)5, enhance luminal clearance of tissue neutrophils by CD55 expression 59 and augment mucosal host defense via expression of anti-microbial proteins 25,26. These pro-resolution mediators can also stimulate the secretion of transforming growth factor (TGF)-β1 and IL-10 that can also dampen inflammation12,107.

For asthma, currently available anti-inflammatory agents [e.g., glucocorticoids (GCs) and the biological therapeutics anti-IgE, IL-5 or IL-13] target pro-inflammatory mediator pathways39. In addition, asthmatic individuals may use non-steroidal anti-inflammatory drugs that inhibit cyclooxygenases (COX) and zileuton that inhibits 5-lipoxygenase (ALOX5). These agents can interfere with inflammation resolution, because these enzymes are also involved in the endogenous formation of pro-resolving mediators43,44,89. Thus, new disease-modifying therapeutic agents are needed that are specifically designed to prevent further inflammation and promote resolution.

Polyunsaturated fatty acid metabolism to pro-resolving mediators

Polyunsaturated fatty acids (PUFAs) are essential nutrients. The ω-6 PUFA, arachidonic acid (AA; 20:4n-6) is incorporated into cellular phospholipids, and upon cell activation, specific phospholipase A2 enzymes release AA from the sn-2 fatty acyl bond of phospholipids. AA can then be converted enzymatically by COX to prostaglandins (PGs), by ALOX5 to leukotrienes (LTs) or, by ALOX5 in collaboration with ALOX12 or ALOX15 to lipoxins (LXs)83,92. The ω-3 PUFAs eicosapentaenoic acid (EPA; 20:5n−3) and docosahexaenoic acid (DHA; 22:6n−3) are enriched in neural and mucosal tissues 14,40. In whole blood from healthy subjects, the total fatty acid pool contains both EPA (~0.5 to 2.8% of total fatty acids) and DHA (~1.3 to 5.0%) 1,47,57,71,74,113. These ω-3 PUFAs are also available at sites of inflammation for enzymatic transformation to resolvins, protectins, maresins and other new families of pro-resolving mediators95,98,101,104,119. These new specialized mediators display receptor-mediated cell type specific actions (Table 1). Because information on these mediators in airway health and asthma is, at this point, limited, we will focus in this review on lipoxin A4 (LXA4), the E-series resolvin termed resolvin E1 (RvE1) and protectin D1 (PD1) and highlight their formation, sites of action and potential anti-inflammatory and pro-resolving actions with particular reference to available information on allergic airway inflammation and asthma.

Table 1.

Cellular actions of pro-resolving mediators

Pro-resolving
mediator
Cell type Action Reference
Lipoxin A4 Eosinophils Inhibits of eosinophil migration 10
T cells Inhibits of TNF release 4
Neutrophils Inhibits PMN chemotaxis 102
Inhibits trans-endothelial migration 34,76
Inhibits trans-epithelial migration 34
Inhibits of superoxide anion generation 64
Inhibits of azurophilic granule release 108
NK cells Inhibits NK cell cytotoxicity 80
Monocytes Increases adhesion and transmigration 67
Inhibits IL-8 released induce by TNF 20
Macrophages Increases engulfment of apoptotic PMN 46
Smooth muscle Inhibits LTC4-initiated migration 77
Endothelium Stimulates prostacyclin release 22
Epithelial cells Increases proliferation after acid injury 18
Inhibits cytokine release 18
Increases intracellular Ca2+ 19

Resolvin E1 T cells Increases CCR5 expression 5
Neutrophils Inhibits of trans-endothelial migration 25

Protectin D1 T cells Increases CCR5 expression 5
Neutrophils Inhibits TNFα and IFNγ release 6
Inhibits PMN trans-migration 12

Lipoxin A4

Lipoxins (LXs) are tri-hydroxy tetraene containing mediators derived from the sequential actions of LOXs (reviewed in 92). Because LX biosynthetic enzymes (i.e., LOXs) are predominately compartmentalized in distinct cell types, LXs are principally generated during heterotypic cell-cell interactions in multicellular host responses, such as inflammation. LXs are structurally and functionally distinct from PGs and LTs. Tissue levels of these pro-phlogistic eicosanoids (PGs and LTs) are prominent during the onset of inflammation while LXs are increased during resolution. LXs have cell type specific actions (Table 1), including inhibition of neutrophil (PMN) and eosinophil trans-endothelial and trans-epithelial migration, and PMN reactive oxygen species generation, and stimulation of monocyte chemotaxis and macrophage engulfment of apoptotic PMNs. LXs also display neuro-modulatory actions at capsaicin-sensitive sensory nerves in the guinea pig70 and can dampen inflammatory pain processing in mice110. In the lung, ALOX15 is a key enzyme for LX generation and is expressed by many cells in the inflamed lung, including bronchial epithelial cells, macrophages and eosinophils 54,66,100,105. ALOX15 converts C20:4 to 15(S)-hydroperoxyeicosatetraenoic acid (15(S)-H(p)ETE) that can be further converted to LXs by ALOX566. Thus, LX biosynthesis can occur when infiltrating PMNs or eosinophils interact with epithelia in inflamed airways62,63. LXs can also be generated in the vasculature during collaborations between leukocyte ALOX5 and platelet ALOX12. Human platelet ALOX12 is a LX synthase that can generate LXs from ALOX5 derived leukotriene A4 (LTA4).

LX epimers are also generated during inflammation and can be increased in the presence of aspirin (acetylsalicylic acid (ASA))33. Acetylation of COX-2 by ASA blocks prostaglandin synthesis; however, the enzyme remains catalytically active, converting AA to 15(R)-hydroxyeicosapentaenoic acid (15(R)-HETE) rather than PG. 15(R)-HETE can serve as a substrate for leukocyte ALOX5 for subsequent transformation to 15-epimer-LXs33. 15(R)-HETE can also be generated by cytochrome P450 metabolism of C20:4 in epithelial cells32. Of interest, statins can also increase 15-epimer-LX production17, including in the airway79.

At sites of inflammation, LXs are rapidly formed and rapidly inactivated. The enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) metabolizes LXs by dehydration to convert LXA4 into 15-oxo-LXA4 96. This conversion inactivates the LXs, as the oxo-LXs no longer display counter-regulatory bioactions. 15-oxo-LXA4 can be further metabolized by eicosanoid oxidorectuctase, which specifically reduces the double bond adjacent to the ketone, and then again by 15-PGDH to a 13,14-dihydro-LXA4. LX metabolism is stereo-specific, so 15-epi-LXs are metabolized less efficiently than LXs, increasing their biological half-life approximately 2-fold and thereby enhancing their ability to evoke bioactions102. Thus, ASA’s unique pharmacological properties arise from both its ability to inhibit pro-inflammatory PG formation and its capacity to generate anti-inflammatory lipid mediators, including the 15-epi-LXs 109. This biosynthetic paradigm for ASA-triggered counter-regulatory lipid mediators from AA is also observed for the ω-3 polyunsaturated fatty acids C20:5 (eicosapentaenoic acid) and C22:6 (docosahexaenoic acid)109.

Lipoxin A4 receptors

LXA4 is an agonist, interacting with specific receptors (termed ALX/FPR2). ALX/FPR2 receptors are 7-transmembrane, G-protein coupled receptors that are expressed on several cell types, including human PMNs, eosinophils, monocytes/macrophages, enterocytes, synovial fibroblasts and airway epithelium30. LXs bind to ALX/FPR2 with high affinity (Kd= 1.7nM) 29. Asthma biologists will recognize that the DP2 receptor can bind both peptide and lipid ligands 52, but ALX/FPR2 was the first receptor described with these properties 97. This dual recognition of structurally distinct ligands is now appreciated to be a more generalized phenomenon30,7. In PMNs, ALX/FPR2 transduces stop signals in part via polyisoprenyl phosphate remodeling 64 and inhibition of leukocyte specific protein (LSP-1) phosphorylation, a downstream regulator of the p38-MAPK cascade75.

Cytokines, including IL-13 and IFN-γ, can induce ALX/FPR2 expression in airway epithelial cells and enterocytes 48 and the ALX/FPR2 promoter was recently characterized106. Lung ALX/FPR2 expression is also induced in vivo in a murine model of allergic airway inflammation63. Transgenic mice that express human ALX/FPR2 receptors have demonstrated the importance of these receptors in regulating experimental inflammation37, including allergic airway responses63.

In addition to ALX/FPR2, additional receptors have been identified for LXA4, including the intracellular aryl hydrocarbon receptor (AhR)88 and CysLT1 receptor9,49. LX signaling via AhR can attenuate IL-12 release from dendritic cells2. LXA4 interaction with CysLT1 receptors antagonizes CysLT binding 49. Together, these findings indicate that LXs can serve as agonists (via ALX and AhR) to transduce cell type specific anti-inflammatory and pro-resolving actions and as antagonists (at CysLT1) to block pro-phlogistic signaling.

Lipoxin A4 in allergic airway responses and asthma

Lipoxins are present in BALFs and sputum in asthma60,78,112. Because LXs can block LTD4-mediated constriction of isolated lung strips in vitro,36 LXA4 was given via nebulizer to asthmatic subjects prior to airway provocation challenges with inhaled LTC4, and LXA4 displayed protective actions in human asthmatic airways31.

Translational research has uncovered a decreased ability to generate LXs in aspirin-exacerbated respiratory disease84 and other forms of severe asthma61,78 (Table 2). Relative to milder asthma, blood from severe asthmatics has a reduced capacity to convert AA to ALOX15 catalyzed products, including 15-HETE and LXA428,61, and levels of LXA4 in bronchoalveolar lavage fluids (BALFs) are markedly decreased in severe compared to non-severe asthma78. LXA4 levels are also lower in supernatants of induced sputum in severe compared to mild asthma112. In addition, lower levels of LXA4 are associated with exercise-induced asthma111. Decreased formation of LXs in uncontrolled asthma has now been identified in culturally distinct populations of adults and children from several countries, including the U.S., Poland, France, Turkey and China (Table 2). Decreased LX generation in uncontrolled asthma is partially explained by dysregulated expression of LX biosynthetic genes that vary by both disease severity and anatomic compartment78. In contrast, generation of the ALOX5 derived products 5-HETE, LTB4 and CysLTs are all increased in asthma, in particular severe asthma61. Peripheral blood LXA4 and the individualized ratio of LXA4/CysLTs correlate with lung function (FEV1 percent predicted values)28,61, suggesting a link between AA metabolism to these bioactive lipids and airflow obstruction in asthma. Lower LX levels have also been described in the airways of patients suffering from cystic fibrosis and interstitial lung disease and appear to play important roles in the pathobiology of these inflammatory lung diseases as well55,58. Together, these results indicate that LXs are generated during airway inflammation and decrements in their generation can contribute to the pathogenesis of asthma.

Table 2.

Uncontrolled asthma – A defect in pro-resolving mediators

Pro-resolving
mediator
Evidence Age Country Reference
Lipoxin A4 Aspirin-tolerant asthmatics generate more lipoxins than aspirin-intolerant asthmatics. Adults Poland 84
Diminished lipoxin biosynthesis in severe asthma Adults USA 61
Severe asthma is associated with a loss of LXA4, an endogenous anti-inflammatory compound. Adults France 112
Lipoxin A4 levels in asthma: relation with disease severity and aspirin sensitivity Adults Turkey 28
Airway lipoxin A4 generation and lipoxin A4 receptor expression are decreased in severe asthma. Adults USA 78
The role of lipoxin A4 in exercise-induced bronchoconstriction in asthma Adults Turkey 111
Corticosteroid suppression of lipoxin A4 and leukotriene B4 from alveolar macrophages in severe asthma. Adults UK 15
Reversed changes of lipoxin A4 and leukotrienes in children with asthma in different severity degree. Children China 118

Protectin D1 Generated in asthma and dampens airway inflammation and hyper-responsiveness. Adults US 65

Novel Omega-3 fatty acid derived lipid mediators

Population surveys report that diets rich in omega-3 fatty acids are associated with lower asthma prevalence90. However, the underlying mechanisms behind this observation remain unclear. Recently, specialized pro-resolving mediators derived from omega-3 fatty acids were identified in self-limited models of acute inflammation and termed resolvins (resolution phase interaction products) 95,101. Similar to LXs, resolvins can be generated via ALOX5 catalyzed reactions during transcellular biosynthesis (reviewed in 92). Based on the omega-3 fatty acid of origin, resolvins are divided into the D-series (docosahexaenoic acid; DHA, C22:6) or E- series (eicosapentaenoic acid; EPA, C20:5). Dietary modification or transgenic expansion of the fat-1 gene (GeneID: 178291 (NC_003282.5)), which encodes a prokaryote omega-3 fatty acid desaturase, can increase resolvin production35,53.

Resolvin E1

Resolvin E1 (RvE1) is present in resolving exudates95 and lung 16. RvE1 transcellular biosynthesis occurs in human vasculature in the presence of ASA with transformation of EPA to 18R-HEPE (18R-hydroxyeicosapentaenoic acid) by ASA-acetylated COX-2 in endothelial cells7. 18R-HEPE is subsequently converted by leukocyte ALOX5 to RvE1 via a 5(6) epoxide-containing intermediate 7,95. RvE1 biosynthesis in humans can be increased by ingestion of ASA (100g) and/or following dietary EPA supplementation (1g) 7. The stereochemical assignment for RvE1 is 5S,12R,18R-trihydroxyeicosa-6Z,8E,10E,14Z,16E-pentaenoic acid 7,95, and structure-activity relationships have established that RvE1’s actions are highly stereoselective92.

Resolvin E1 Receptors

Receptors RvE1 have been recently identified7,8. RvE1 interacts with the G-protein coupled receptor CMKLR1 as an agonist. CMKLR1 (aka ChemR23) was first identified as a receptor for the peptide chemerin. Thus, like ALX/FPR2, this receptor also recognizes both lipid and peptide ligands. CMKLR1 is expressed on monocytes/macrophages and plasmacytoid dendritic cells 27,81,115. Human CMKLR1 binds RvE1 with high affinity (Kd = 11.3 ± 5.4 nM) 7. RvE1 also interacts with a second G-protein coupled receptor: leukotriene B4 receptor 1 (BLT1) 8. In humans, BLT1 receptors are expressed by neutrophils, eosinophils, monocytes/macrophages, mast cells, dendritic cells, and effector T cells 120. RvE1 effectively competes with LTB4 for BLT1 binding to functionally antagonize LTB4-mediated responses8. RvE1’s affinity for BLT1 (Kd = 45nm) is relatively lower than for CMKLR1 and in addition to LTB4 antagonism, RvE1 can serve as a partial agonist for BLT1 signaling.

Resolvin E1 in allergic airway responses and asthma

Resolvin E1 has potent anti-inflammatory and pro-resolving actions in a number of murine models of inflammation and is bioactive in very low concentrations (nanomolar to picomolar) in vivo and in vitro95. RvE1 dampens the development and promotes the resolution of allergic airway responses in a murine experimental model of asthma3,50,51. In nanogram quantities, intravenous administration of RvE1 decreases eosinophil and lymphocyte accumulation and airway mucous metaplasia and improves airway hyper-responsiveness to inhaled methacholine51. To promote resolution of inflammation, RvE1 decreases IL-6, IL-17 and IL-23 in the lung while increasing IFN-γ and LXA4 formation51. RvE1’s actions were additive with those of LXs, as these pro-resolving mediators both regulate IL-17. Of interest, only RvE1, and not LXA4, regulated IL-23 and IFNγ levels51. Together, these findings support the presence of independent pro-resolving signalling 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 of this asthma model3. Administration with RvE1 concurrent with allergen challenge can prevent the development of bronchial hyper-responsiveness, mucous metaplasia, eosinophil accumulation and T-helper type 2 (Th2) cytokine mediator release [e.g. IL-13].3 Because immune responses are pathologically sustained in asthma, models of self-limited allergic airway responses have been used to determine mechanisms for down-regulating adaptive immune responses in health with the goal of establishing the critical checkpoints for maintaining lung tissue homeostasis. In a murine model of asthma exacerbation with self-limited allergic airway inflammation, natural killer (NK) cell phenotype and trafficking to draining mediastinal lymph nodes is temporally regulated during resolution in a CXCL9-CXCR3 (ligand-receptor) dependent manner50. Depletion of NK cells disrupts the endogenous resolution program, leading to delayed resolution of airway eosinophils and antigen specific CD4+ T cells, increased LTB4 and interleukin-23, and decreased LXA450. In addition, NK cells express CMKLR1 and RvE1 mediated resolution is markedly inhibited in this setting by NK cell depletion. These findings indicate new functions for NK cells in promoting resolution of adaptive immune responses and suggest that NK cells are targets for specialized pro-resolving mediators for clearance of activated T cells from inflamed lung. No published reports are currently available for Resolvin E1 in human asthma.

Protectin D1

DHA is also present in resolving exudates and DHA is converted into D-Series resolvins101. Of interest, the DHA metabolome includes several additional classes of pro-resolving mediators, including protectins and maresins98,104. Mucosal surfaces, including the airway, are enriched with DHA41, and airway mucosal epithelial cells from individuals with asthma or cystic fibrosis have depleted stores of DHA compared with healthy control subjects 41. This section will focus on protectin D1 (PD1). Roles for D-Series resolvins, maresins and other DHA derived mediators in asthma is the subject ongoing research; however, there is currently only limited information on these compounds in asthma.

At sites of inflammation, DHA is rapidly converted by LOX activity to a 17S- hydroperoxy-containing intermediate and via an epoxide containing intermediate to protectins, such as PD1 (10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid). 99 When produced in neural systems this bioactive mediator is coined neuroprotectin D1 NPD1/PD1 (for detailed reviews, see 13,92,94). NPD1/PD1 is distinguished from D-Series resolvins by the presence of a conjugated triene and two alcohol groups in a specific chirality required for potent bioactions. Structure-activity relationships suggest that PD1’s actions are receptor-mediated in human leukocytes69; however, the PD1 receptor has yet to be molecularly characterized.

Protectin D1 in allergic airway responses and asthma

DHA can be delivered during the early stages of an acute inflammatory response in albumin-rich edema fluid56. It is noteworthy that the cellular components of the airway mucosa are enriched with DHA 40 and both 17S-HDHA (17S-hydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid) and PD1 are generated in human airways65. Of interest, PD1 levels decrease during acute exacerbations of asthma (Table 2) and PD1 mediates bronchoprotective actions in a murine experimental model of allergic asthma in vivo 65. Using ovalbumin-sensitized mice, PD1 (2 - 200 ng) given by the intravenous route prior to aeroallergen challenge markedly suppressed bronchial hyper-responsiveness, mucous metaplasia, pulmonary eosinophil accumulation and pro-inflammatory cytokine and lipid mediator release. PD1 also mediates pro-resolving actions on established pulmonary inflammation when given after allergen challenge, leading to significantly expedited resolution of allergic airway inflammation. 65 In a head to head comparison with a lipoxin stable analog and RvE1 in acute, self-limited murine peritonitis, PD1 displayed the lowest Ψmax (maximum number of infiltrating leukocytes) and Tmax (time to resolve) and the shortest RI (resolution interval, time taken from maximal leukocyte infiltration to 50% of maximum), indicative of PD1’s potent bioactons 12.

Summary and Conclusions

The resolution of inflammation is an integral and natural part of the physiological response to tissue injury, infection and allergen or other noxious stimuli. Resolution is an active process with highly regulated cellular and biochemical events. Several discrete families of natural small molecules have recently been uncovered using a lipidomics approach. Select members are pro-resolving and stimulate tissue catabasis. These include resolvins, protectins and most recently maresins. Insights into these chemical mediators and their signaling pathways provide an opportunity to develop agonists of resolution as a potential novel class of therapeutics. The chronic and uncontrolled airway inflammation that characterizes asthma, especially severe variants of the disease, appears to result in part from a defect in resolution. It is notable that in uncontrolled asthma there is also a defect in the generation of specialized pro-resolving mediators, including LXA4 and PD1. Thus, bioactive stable analog mimetics of these mediators that can harness endogenous resolution mechanisms for inflammation may offer new therapeutic strategies for asthma and airway inflammation associated diseases.

Key Points.

  1. Resolution of inflammation is an active process with specific cellular and molecular events.

  2. Specialized pro-resolving mediators are enzymatically derived from polyunsaturated fatty acids, including the omega-3 fatty acids EPA and DHA.

  3. Pro-resolving mediators interact with specific receptors to regulate allergic airway responses and asthma.

  4. Uncontrolled asthma is associated with a defect in pro-resolving mediator signaling.

ACKNOWLEDGMENTS

Funding Sources:

Dr Levy: National Institutes of Health, Lovelace Respiratory Research Institute

Dr Vachier: Med Bio Med

Dr Serhan: National Institutes of Health

This research was supported in part by the US National Institutes of Health grants P01-GM095467, U10-HL109172 and P50-HL107166.

List of Abbreviations

12-LO

12-lipoxygenase

15(R)-HETE

15(R)-hydroxyeicosatetraenoic acid

15(S)-H(p)ETE)

15(S)-hydroperoxyeicosatetraenoic acid

15-LO

15-lipoxygenase

15-PGDH

15-hydroxyprostaglandin dehydrogenase

5-LO

5-lipoxygenase

AhR

aryl hydrocarbon receptor

ASA

acetylsalicylic acid

ATLa

aspirin-triggered lipoxin analog

BALF

bronchoalveolar lavage fluid

C20:4

arachidonic acid

C20:5

eicosapentaenoic acid (EPA)

C22:6

docosahexaenoic acid (DHA)

COX-2

cyclooxygenase-2

CysLTs

cysteinyl leukotrienes

LSP-1

leukocyte specific protein-1

LTA4

leukotriene A4

LTA4H

leukotriene A4 hydrolase

LTC4

leukotriene C4

LXA4

lipoxin A4

PD1

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

PGs

prostaglandins

PMNs

polymorphonuclear leukocytes

Rv

resolvin

RvD1

Resolvin D1 (7S,8R,17S trihydroxydocosa-4Z,9E,11E,13Z,15E,19Z-hexaenoic acid)

RvE1

Resolvin E1 (5S,12R,18R-trihydroxyeicosa-6Z,8E,10E,14Z,16E-pentaenoic acid)

SPM

specialized pro-resolving mediators

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.

DISCLOSURES

Conflict of Interest:

Dr Levy: Consultant for Resolvyx Pharmaceuticals; Patents licensed to Bayer Healthcare and Resolvyx; Equity in Resolvyx Pharmaceuticals. BDL’s interests were reviewed and are managed by the Brigham and Women’s Hospital and Partners HealthCare in accordance with their conflict of interest policies.

Dr Vachier: No conflict of Interest

Dr. Serhan: CNS is an inventor on patents [resolvins] and pro-resolving mediators and related compounds assigned to BWH and licensed to Resolvyx Pharmaceuticals. CNS is a scientific founder of Resolvyx Pharmaceuticals and owns equity in the company. CNS’ interests were reviewed and are managed by the Brigham and Women’s Hospital and Partners HealthCare in accordance with their conflict of interest policies.

Bibliography

  • 1.Albert CM, Campos H, Stampfer MJ, et al. Blood levels of long-chain n-3 fatty acids and the risk of sudden death. N Engl J Med. 2002;346:1113. doi: 10.1056/NEJMoa012918. [DOI] [PubMed] [Google Scholar]
  • 2.Aliberti J, Hieny S, Reis e Sousa C, et al. Lipoxin-mediated inhibition of IL-12 production by DCs: a mechanism for regulation of microbial immunity. Nat Immunol. 2002;3:76. doi: 10.1038/ni745. [DOI] [PubMed] [Google Scholar]
  • 3.Aoki H, Hisada T, Ishizuka T, et al. Resolvin E1 dampens airway inflammation and hyperresponsiveness in a murine model of asthma. Biochem Biophys Res Commun. 2008;367:509. doi: 10.1016/j.bbrc.2008.01.012. [DOI] [PubMed] [Google Scholar]
  • 4.Ariel A, Chiang N, Arita M, et al. Aspirin-triggered lipoxin A4 and B4 analogs block extracellular signal-regulated kinase-dependent TNF-alpha secretion from human T cells. J Immunol. 2003;170:6266. doi: 10.4049/jimmunol.170.12.6266. [DOI] [PubMed] [Google Scholar]
  • 5.Ariel A, Fredman G, Sun YP, et al. Apoptotic neutrophils and T cells sequester chemokines during immune response resolution through modulation of CCR5 expression. Nat Immunol. 2006;7:1209. doi: 10.1038/ni1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ariel A, Li PL, Wang W, et al. The docosatriene protectin D1 is produced by TH2 skewing and promotes human T cell apoptosis via lipid raft clustering. J Biol Chem. 2005;280:43079. doi: 10.1074/jbc.M509796200. [DOI] [PubMed] [Google Scholar]
  • 7.Arita M, Bianchini F, Aliberti J, et al. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. Journal of Experimental Medicine. 2005;201:713. doi: 10.1084/jem.20042031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Arita M, Ohira T, Sun YP, et al. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. Journal of Immunology. 2007;178:3912. doi: 10.4049/jimmunol.178.6.3912. [DOI] [PubMed] [Google Scholar]
  • 9.Badr KF, DeBoer DK, Schwartzberg M, et al. Lipoxin A4 antagonizes cellular and in vivo actions of leukotriene D4 in rat glomerular mesangial cells: evidence for competition at a common receptor. Proceedings of the National Academy of Sciences of the United States of America. 1989;86:3438. doi: 10.1073/pnas.86.9.3438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bandeira-Melo C, Bozza PT, Diaz BL, et al. Cutting edge: lipoxin (LX) A4 and aspirintriggered 15-epi-LXA4 block allergen-induced eosinophil trafficking. J Immunol. 2000;164:2267. doi: 10.4049/jimmunol.164.5.2267. [DOI] [PubMed] [Google Scholar]
  • 11.Bandeira-Melo C, Serra MF, Diaz BL, et al. Cyclooxygenase-2-derived prostaglandin E2 and lipoxin A4 accelerate resolution of allergic edema in Angiostrongylus costaricensisinfected rats: relationship with concurrent eosinophilia. Journal of Immunology. 2000;164:1029. doi: 10.4049/jimmunol.164.2.1029. [DOI] [PubMed] [Google Scholar]
  • 12.Bannenberg GL, Chiang N, Ariel A, et al. Molecular circuits of resolution: formation and actions of resolvins and protectins.[erratum appears in J Immunol. 2005 May 1;174(9):5884] Journal of Immunology. 2005;174:4345. doi: 10.4049/jimmunol.174.7.4345. [DOI] [PubMed] [Google Scholar]
  • 13.Bazan NG. Neuroprotectin D1-mediated anti-inflammatory and survival signaling in stroke, retinal degenerations, and Alzheimer's disease. J Lipid Res. 2009;50:S400. doi: 10.1194/jlr.R800068-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bazan NG. Synaptic lipid signaling: significance of polyunsaturated fatty acids and platelet-activating factor. J Lipid Res. 2003;44:2221. doi: 10.1194/jlr.R300013-JLR200. [DOI] [PubMed] [Google Scholar]
  • 15.Bhavsar PK, Levy BD, Hew MJ, et al. Corticosteroid suppression of lipoxin A4 and leukotriene B4 from alveolar macrophages in severe asthma. Respir. 2010;11:71. doi: 10.1186/1465-9921-11-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bilal S, Haworth O, Wu L, et al. Fat-1 transgenic mice with elevated omega-3 fatty acids are protected from allergic airway responses. Biochimie Biophysica Acta. 2011;1812:1164. doi: 10.1016/j.bbadis.2011.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Birnbaum Y, Ye Y, Lin Y, et al. Augmentation of myocardial production of 15-epi-lipoxin-a4 by pioglitazone and atorvastatin in the rat. Circulation. 2006;114:929. doi: 10.1161/CIRCULATIONAHA.106.629907. [DOI] [PubMed] [Google Scholar]
  • 18.Bonnans C, Fukunaga K, Levy MA, et al. Lipoxin A(4) regulates bronchial epithelial cell responses to acid injury. Am J Pathol. 2006;168:1064. doi: 10.2353/ajpath.2006.051056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bonnans C, Mainprice B, Chanez P, et al. Lipoxin A4 stimulates a cytosolic Ca2+ increase in human bronchial epithelium. Journal of Biological Chemistry. 2003;278:10879. doi: 10.1074/jbc.M210294200. [DOI] [PubMed] [Google Scholar]
  • 20.Bonnans C, Vachier I, Chavis C, et al. Lipoxins are potential endogenous antiinflammatory mediators in asthma. American Journal of Respiratory & Critical Care Medicine. 2002;165:1531. doi: 10.1164/rccm.200201-053OC. [DOI] [PubMed] [Google Scholar]
  • 21.Bousquet J, Jeffery PK, Busse WW, et al. Asthma. From bronchoconstriction to airways inflammation and remodeling. American Journal of Respiratory and Critical Care Medicine. 2000;161:1720. doi: 10.1164/ajrccm.161.5.9903102. [DOI] [PubMed] [Google Scholar]
  • 22.Brezinski ME, Gimbrone MA, Jr, Nicolaou KC, et al. Lipoxins stimulate prostacyclin generation by human endothelial cells. FEBS Lett. 1989;245:167. doi: 10.1016/0014-5793(89)80214-5. [DOI] [PubMed] [Google Scholar]
  • 23.Burr GOBM. A new deficiency disease produced by the rigid exclusion of fat from the diet. Biol Chem. 1929;82:345. doi: 10.1111/j.1753-4887.1973.tb06008.x. [DOI] [PubMed] [Google Scholar]
  • 24.Busse WW, Lemanske RF., Jr Asthma. New England Journal of Medicine. 2001;344:350. doi: 10.1056/NEJM200102013440507. [DOI] [PubMed] [Google Scholar]
  • 25.Campbell EL, Louis NA, Tomassetti SE, et al. Resolvin E1 promotes mucosal surface clearance of neutrophils: a new paradigm for inflammatory resolution. FASEB J. 2007;21:3162. doi: 10.1096/fj.07-8473com. [DOI] [PubMed] [Google Scholar]
  • 26.Canny G, Levy O, Furuta GT, et al. Lipid mediator-induced expression of bactericidal/ permeability-increasing protein (BPI) in human mucosal epithelia. Proc Natl Acad Sci U S A. 2002;99:3902. doi: 10.1073/pnas.052533799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cash JL, Hart R, Russ A, et al. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J Exp Med. 2008;205:767. doi: 10.1084/jem.20071601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Celik GE, Erkekol FO, Misirligil Z, et al. Lipoxin A4 levels in asthma: relation with disease severity and aspirin sensitivity. Clinical & Experimental Allergy. 2007;37:1494. doi: 10.1111/j.1365-2222.2007.02806.x. [DOI] [PubMed] [Google Scholar]
  • 29.Chiang N, Fierro IM, Gronert K, et al. Activation of lipoxin A(4) receptors by aspirin-triggered lipoxins and select peptides evokes ligand-specific responses in inflammation. J Exp Med. 2000;191:1197. doi: 10.1084/jem.191.7.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chiang N, Serhan CN, Dahlen SE, et al. The lipoxin receptor ALX: potent ligand-specific and stereoselective actions in vivo. Pharmacol Rev. 2006;58:463. doi: 10.1124/pr.58.3.4. [DOI] [PubMed] [Google Scholar]
  • 31.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. doi: 10.1164/ajrccm/145.6.1281. [DOI] [PubMed] [Google Scholar]
  • 32.Claria J, Lee MH, Serhan CN. Aspirin-triggered lipoxins (15-epi-LX) are generated by the human lung adenocarcinoma cell line (A549)-neutrophil interactions and are potent inhibitors of cell proliferation. Molecular Medicine. 1996;2:583. [PMC free article] [PubMed] [Google Scholar]
  • 33.Claria J, Serhan CN. Aspirin triggers previously undescribed bioactive eicosanoids by human endothelial cell-leukocyte interactions. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:9475. doi: 10.1073/pnas.92.21.9475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Colgan SP, Serhan CN, Parkos CA, et al. Lipoxin A4 modulates transmigration of human neutrophils across intestinal epithelial monolayers. J Clin Invest. 1993;92:75. doi: 10.1172/JCI116601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Connor KM, SanGiovanni JP, Lofqvist C, et al. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med. 2007;13:868. doi: 10.1038/nm1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Dahlen SE, Franzen L, Raud J, et al. Actions of lipoxin A4 and related compounds in smooth muscle preparations and on the microcirculation in vivo. Adv Exp Med Biol. 1988;229:107. doi: 10.1007/978-1-4757-0937-7_9. [DOI] [PubMed] [Google Scholar]
  • 37.Devchand PR, Arita M, Hong S, et al. Human ALX receptor regulates neutrophil recruitment in transgenic mice: roles in inflammation and host defense.[erratum appears in FASEB J. 2003 Jun;17(9):4] FASEB Journal. 2003;17:652. doi: 10.1096/fj.02-0770com. [DOI] [PubMed] [Google Scholar]
  • 38.Fadok VA, Bratton DL, Konowal A, et al. 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. doi: 10.1172/JCI1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Fanta CH. Asthma. N Engl J Med. 2009;360:1002. doi: 10.1056/NEJMra0804579. [DOI] [PubMed] [Google Scholar]
  • 40.Freedman SD, Blanco PG, Zaman MM, et al. Association of cystic fibrosis with abnormalities in fatty acid metabolism. N Engl J Med. 2004;350:560. doi: 10.1056/NEJMoa021218. [DOI] [PubMed] [Google Scholar]
  • 41.Freedman SD, Blanco PG, Zaman MM, et al. Association of cystic fibrosis with abnormalities in fatty acid metabolism.[see comment] New England Journal of Medicine. 2004;350:560. doi: 10.1056/NEJMoa021218. [DOI] [PubMed] [Google Scholar]
  • 42.Freire-de-Lima CG, Xiao YQ, Gardai SJ, et al. 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. doi: 10.1074/jbc.M605146200. [DOI] [PubMed] [Google Scholar]
  • 43.Fukunaga K, Kohli P, Bonnans C, et al. Cyclooxygenase 2 plays a pivotal role in the resolution of acute lung injury. Journal of Immunology. 2005;174:5033. doi: 10.4049/jimmunol.174.8.5033. [DOI] [PubMed] [Google Scholar]
  • 44.Gilroy DW, Colville-Nash PR, Willis D, et al. Inducible cyclooxygenase may have anti-inflammatory properties [see comments] Nature Medicine. 1999;5:698. doi: 10.1038/9550. [DOI] [PubMed] [Google Scholar]
  • 45.Gilroy DW, Lawrence T, Perretti M, et al. Inflammatory resolution: new opportunities for drug discovery. Nature Reviews. Drug Discovery. 2004;3:401. doi: 10.1038/nrd1383. [DOI] [PubMed] [Google Scholar]
  • 46.Godson C, Mitchell S, Harvey K, et al. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. Journal of Immunology. 2000;164:1663. doi: 10.4049/jimmunol.164.4.1663. [DOI] [PubMed] [Google Scholar]
  • 47.Gong J, Rosner B, Rees DG, et al. Plasma docosahexaenoic acid levels in various genetic forms of retinitis pigmentosa. Invest Ophthalmol Vis Sci. 1992;33:2596. [PubMed] [Google Scholar]
  • 48.Gronert K, Gewirtz A, Madara JL, et al. Identification of a human enterocyte lipoxin A4 receptor that is regulated by interleukin (IL)-13 and interferon gamma and inhibits tumor necrosis factor alpha-induced IL-8 release. J Exp Med. 1998;187:1285. doi: 10.1084/jem.187.8.1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gronert K, Martinsson-Niskanen T, Ravasi S, et al. Selectivity of recombinant human leukotriene D(4), leukotriene B(4), and lipoxin A(4) receptors with aspirin-triggered 15-epi-LXA(4) and regulation of vascular and inflammatory responses. Am J Pathol. 2001;158:3. doi: 10.1016/S0002-9440(10)63937-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Haworth O, Cernadas M, Levy BD. NK cells are effectors for resolvin E1 in the timely resolution of allergic airway inflammation. Journal of Immunology. 2011;186:6129. doi: 10.4049/jimmunol.1004007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Haworth O, Cernadas M, Yang R, et al. Resolvin E1 regulates interleukin 23, interferon-gamma and lipoxin A4 to promote the resolution of allergic airway inflammation. Nature Immunology. 2008;9:873. doi: 10.1038/ni.1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hirai H, Tanaka K, Yoshie O, et al. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. J Exp Med. 2001;193:255. doi: 10.1084/jem.193.2.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hudert CA, Weylandt KH, Lu Y, et al. Transgenic mice rich in endogenous omega-3 fatty acids are protected from colitis. Proc Natl Acad Sci U S A. 2006;103:11276. doi: 10.1073/pnas.0601280103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hunter JA, Finkbeiner WE, Nadel JA, et al. Predominant generation of 15-lipoxygenase metabolites of arachidonic acid by epithelial cells from human trachea. Proceedings of the National Academy of Sciences of the United States of America. 1985;82:4633. doi: 10.1073/pnas.82.14.4633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Karp CL, Flick LM, Park KW, et al. Defective lipoxin-mediated anti-inflammatory activity in the cystic fibrosis airway. Nat Immunol. 2004;5:388. doi: 10.1038/ni1056. [DOI] [PubMed] [Google Scholar]
  • 56.Kasuga K, Yang R, Porter TF, et al. Rapid appearance of resolvin precursors in inflammatory exudates: novel mechanisms in resolution. J Immunol. 2008;181:8677. doi: 10.4049/jimmunol.181.12.8677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kew S, Mesa MD, Tricon S, et al. 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. doi: 10.1093/ajcn/79.4.674. [DOI] [PubMed] [Google Scholar]
  • 58.Kowal-Bielecka O, Kowal K, Distler O, et al. Cyclooxygenase- and lipoxygenase-derived eicosanoids in bronchoalveolar lavage fluid from patients with scleroderma lung disease: an imbalance between proinflammatory and antiinflammatory lipid mediators. Arthritis Rheum. 2005;52:3783. doi: 10.1002/art.21432. [DOI] [PubMed] [Google Scholar]
  • 59.Lawrence DW, Bruyninckx WJ, Louis NA, et al. Antiadhesive role of apical decay-accelerating factor (CD55) in human neutrophil transmigration across mucosal epithelia. J Exp Med. 2003;198:999. doi: 10.1084/jem.20030380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lee TH, Crea AE, Gant V, et al. Identification of lipoxin A4 and its relationship to the sulfidopeptide leukotrienes C4, D4, and E4 in the bronchoalveolar lavage fluids obtained from patients with selected pulmonary diseases. American Review of Respiratory Disease. 1990;141:1453. doi: 10.1164/ajrccm/141.6.1453. [DOI] [PubMed] [Google Scholar]
  • 61.Levy BD, Bonnans C, Silverman ES, et al. Diminished lipoxin biosynthesis in severe asthma. American Journal of Respiratory & Critical Care Medicine. 2005;172:824. doi: 10.1164/rccm.200410-1413OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Levy BD, Clish CB, Schmidt B, et al. Lipid mediator class switching during acute inflammation: signals in resolution. Nature Immunology. 2001;2:612. doi: 10.1038/89759. [DOI] [PubMed] [Google Scholar]
  • 63.Levy BD, De Sanctis GT, Devchand PR, et al. Multi-pronged inhibition of airway hyperresponsiveness and inflammation by lipoxin A(4).[see comment] Nature Medicine. 2002;8:1018. doi: 10.1038/nm748. [DOI] [PubMed] [Google Scholar]
  • 64.Levy BD, Fokin VV, Clark JM, et al. Polyisoprenyl phosphate (PIPP) signaling regulates phospholipase D activity: a 'stop' signaling switch for aspirin-triggered lipoxin A4. Faseb J. 1999;13:903. doi: 10.1096/fasebj.13.8.903. [DOI] [PubMed] [Google Scholar]
  • 65.Levy BD, Kohli P, Gotlinger K, et al. Protectin D1 is generated in asthma and dampens airway inflammation and hyperresponsiveness. Journal of Immunology. 2007;178:496. doi: 10.4049/jimmunol.178.1.496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Levy BD, Romano M, Chapman HA, et al. Human alveolar macrophages have 15-lipoxygenase and generate 15(S)-hydroxy-5,8,11-cis-13-trans-eicosatetraenoic acid and lipoxins. Journal of Clinical Investigation. 1993;92:1572. doi: 10.1172/JCI116738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.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. doi: 10.1084/jem.183.1.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Majno G. Cells, Tissues and Disease:Principles of General Pathology. Cambridge, MA: Blackwell; 1996. [Google Scholar]
  • 69.Marcheselli VL, Mukherjee PK, Arita M, et al. Neuroprotectin D1/protectin D1 stereoselective and specific binding with human retinal pigment epithelial cells and neutrophils. Prostaglandins Leukot Essent Fatty Acids. 2010;82:27. doi: 10.1016/j.plefa.2009.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Meini S, Evangelista S, Geppetti P, et al. Pharmacologic and neurochemical evidence for the activation of capsaicin-sensitive sensory nerves by lipoxin A4 in guinea pig bronchus. Am Rev Respir Dis. 1992;146:930. doi: 10.1164/ajrccm/146.4.930. [DOI] [PubMed] [Google Scholar]
  • 71.Miles EA, Noakes PS, Kremmyda LS, et al. The Salmon in Pregnancy Study: study design, subject characteristics, maternal fish and marine n-3 fatty acid intake, and marine n-3 fatty acid status in maternal and umbilical cord blood. Am J Clin Nutr. 2011;94:1986S. doi: 10.3945/ajcn.110.001636. [DOI] [PubMed] [Google Scholar]
  • 72.Moore WC, Bleecker ER, Curran-Everett D, et al. Characterization of the severe asthma phenotype by the National Heart, Lung, and Blood Institute's Severe Asthma Research Program. Journal of Allergy & Clinical Immunology. 2007;119:405. doi: 10.1016/j.jaci.2006.11.639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Nathan C, Ding A. Nonresolving inflammation. Cell. 140:871. doi: 10.1016/j.cell.2010.02.029. [DOI] [PubMed] [Google Scholar]
  • 74.Newcomer LM, King IB, Wicklund KG, et al. The association of fatty acids with prostate cancer risk. Prostate. 2001;47:262. doi: 10.1002/pros.1070. [DOI] [PubMed] [Google Scholar]
  • 75.Ohira T, Bannenberg G, Arita M, et al. A stable aspirin-triggered lipoxin A4 analog blocks phosphorylation of leukocyte-specific protein 1 in human neutrophils. J Immunol. 2004;173:2091. doi: 10.4049/jimmunol.173.3.2091. [DOI] [PubMed] [Google Scholar]
  • 76.Papayianni A, Serhan CN, Brady HR. Lipoxin A4 and B4 inhibit leukotriene-stimulated interactions of human neutrophils and endothelial cells. J Immunol. 1996;156:2264. [PubMed] [Google Scholar]
  • 77.Parameswaran K, Radford K, Fanat A, et al. Modulation of human airway smooth muscle migration by lipid mediators and Th-2 cytokines. American Journal of Respiratory Cell & Molecular Biology. 2007;37:240. doi: 10.1165/rcmb.2006-0172OC. [DOI] [PubMed] [Google Scholar]
  • 78.Planaguma A, Kazani S, Marigowda G, et al. Airway lipoxin A4 generation and lipoxin A4 receptor expression are decreased in severe asthma. American Journal of Respiratory & Critical Care Medicine. 2008;178:574. doi: 10.1164/rccm.200801-061OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Planaguma A, Pfeffer MA, Rubin G, et al. Lovastatin decreases acute mucosal inflammation via 15-epi-lipoxin A4. Mucosal immunology. 2010;3:270. doi: 10.1038/mi.2009.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ramstedt U, Serhan CN, Nicolaou KC, et al. Lipoxin A-induced inhibition of human natural killer cell cytotoxicity: studies on stereospecificity of inhibition and mode of action. J Immunol. 1987;138:266. [PubMed] [Google Scholar]
  • 81.Samson M, Edinger AL, Stordeur P, et al. 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. doi: 10.1002/(SICI)1521-4141(199805)28:05<1689::AID-IMMU1689>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
  • 82.Samuelsson B. Les Prix Nobel: Nobel Prizes, Presentations, Biographies and Lectures. Stockholm: Almqvist & Wiksell; 1982. From studies of biochemical mechanisms to novel biological mediators: prostaglandin endoperoxides, thromboxanes and leukotrienes; p. 153. [DOI] [PubMed] [Google Scholar]
  • 83.Samuelsson B, Dahlen SE, Lindgren JA, et al. Leukotrienes and lipoxins: structures, biosynthesis, and biological effects. Science. 1987;237:1171. doi: 10.1126/science.2820055. [DOI] [PubMed] [Google Scholar]
  • 84.Sanak M, Levy BD, Clish CB, et al. Aspirin-tolerant asthmatics generate more lipoxins than aspirin-intolerant asthmatics. European Respiratory Journal. 2000;16:44. doi: 10.1034/j.1399-3003.2000.16a08.x. [DOI] [PubMed] [Google Scholar]
  • 85.Savill J. Apoptosis. Phagocytic docking without shocking. Nature. 1998;392:442. doi: 10.1038/33025. [DOI] [PubMed] [Google Scholar]
  • 86.Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature. 2000;407:784. doi: 10.1038/35037722. [DOI] [PubMed] [Google Scholar]
  • 87.Savill JS, Wyllie AH, Henson JE, et al. 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. doi: 10.1172/JCI113970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Schaldach CM, Riby J, Bjeldanes LF. Lipoxin A4: a new class of ligand for the Ah receptor. Biochemistry. 1999;38:7594. doi: 10.1021/bi982861e. [DOI] [PubMed] [Google Scholar]
  • 89.Schwab JM, Chiang N, Arita M, et al. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature. 2007;447:869. doi: 10.1038/nature05877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Schwartz J, Weiss ST. The relationship of dietary fish intake to level of pulmonary function in the first National Health and Nutrition Survey (NHANES I) Eur Respir J. 1994;7:1821. doi: 10.1183/09031936.94.07101821. [DOI] [PubMed] [Google Scholar]
  • 91.Serhan CN. The resolution of inflammation: the devil in the flask and in the details. FASEB J. 2011;25:1441. doi: 10.1096/fj.11-0502ufm. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Serhan CN. Resolution phase of inflammation: novel endogenous anti-inflammatory and proresolving lipid mediators and pathways. Annual Review of Immunology. 2007;25:101. doi: 10.1146/annurev.immunol.25.022106.141647. [DOI] [PubMed] [Google Scholar]
  • 93.Serhan CN, Brain SD, Buckley CD, et al. Resolution of inflammation: state of the art, definitions and terms. FASEB J. 2007;21:325. doi: 10.1096/fj.06-7227rev. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nature Reviews. Immunology. 2008;8:349. doi: 10.1038/nri2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Serhan CN, Clish CB, Brannon J, et al. 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. doi: 10.1084/jem.192.8.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Serhan CN, Fiore S, Brezinski DA, et al. Lipoxin A4 metabolism by differentiated HL-60 cells and human monocytes: conversion to novel 15-oxo and dihydro products. Biochemistry. 1993;32:6313. doi: 10.1021/bi00076a002. [DOI] [PubMed] [Google Scholar]
  • 97.Serhan CN, Fiore S, Levy BD. Cell-cell interactions in lipoxin generation and characterization of lipoxin A4 receptors. Annals of the New York Academy of Sciences. 1994;744:166. doi: 10.1111/j.1749-6632.1994.tb52734.x. [DOI] [PubMed] [Google Scholar]
  • 98.Serhan CN, Fredman G, Yang R, et al. Novel proresolving aspirin-triggered DHA pathway. Chemical Biology. 2011;18:976. doi: 10.1016/j.chembiol.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Serhan CN, Gotlinger K, Hong S, et al. Anti-inflammatory actions of neuroprotectin D1/protectin D1 and its natural stereoisomers: assignments of dihydroxy-containing docosatrienes. J Immunol. 2006;176:1848. doi: 10.4049/jimmunol.176.3.1848. [DOI] [PubMed] [Google Scholar]
  • 100.Serhan CN, Hirsch U, Palmblad J, et al. Formation of lipoxin A by granulocytes from eosinophilic donors. FEBS Lett. 1987;217:242. doi: 10.1016/0014-5793(87)80671-3. [DOI] [PubMed] [Google Scholar]
  • 101.Serhan CN, Hong S, Gronert K, et al. 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. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Serhan CN, Maddox JF, Petasis NA, et al. Design of lipoxin A4 stable analogs that block transmigration and adhesion of human neutrophils. Biochemistry. 1995;34:14609. doi: 10.1021/bi00044a041. [DOI] [PubMed] [Google Scholar]
  • 103.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nature Immunology. 2005;6:1191. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 104.Serhan CN, Yang R, Martinod K, et al. Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions. Journal of Experimental Medicine. 2009;206:15. doi: 10.1084/jem.20081880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Shannon VR, Chanez P, Bousquet J, et al. Histochemical evidence for induction of arachidonate 15-lipoxygenase in airway disease. American Review of Respiratory Disease. 1993;147:1024. doi: 10.1164/ajrccm/147.4.1024. [DOI] [PubMed] [Google Scholar]
  • 106.Simiele F, Recchiuti A, Mattoscio D, et al. Transcriptional regulation of the human FPR2/ALX gene: evidence of a heritable genetic variant that impairs promoter activity. FASEB J. 2011 doi: 10.1096/fj.11-198069. in press. [DOI] [PubMed] [Google Scholar]
  • 107.Souza DG, Fagundes CT, Amaral FA, et al. 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. doi: 10.4049/jimmunol.179.12.8533. [DOI] [PubMed] [Google Scholar]
  • 108.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-Lleucyl-L-phenylalanine. Allergy. 1994;49:230. doi: 10.1111/j.1398-9995.1994.tb02654.x. [DOI] [PubMed] [Google Scholar]
  • 109.Spite M, Serhan CN. Novel lipid mediators promote resolution of acute inflammation: impact of aspirin and statins. Circ Res. 2010;107:1170. doi: 10.1161/CIRCRESAHA.110.223883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Svensson CI, Zattoni M, Serhan CN. Lipoxins and aspirin-triggered lipoxin inhibit inflammatory pain processing. J Exp Med. 2007;204:245. doi: 10.1084/jem.20061826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tahan F, Saraymen R, Gumus H. The role of lipoxin A4 in exercise-induced bronchoconstriction in asthma. Journal of Asthma. 2008;45:161. doi: 10.1080/02770900701847068. [DOI] [PubMed] [Google Scholar]
  • 112.Vachier I, Bonnans C, Chavis C, et al. Severe asthma is associated with a loss of LX4, an endogenous anti-inflammatory compound. J Allergy Clin Immunol. 2005;115:55. doi: 10.1016/j.jaci.2004.09.038. [DOI] [PubMed] [Google Scholar]
  • 113.Wakai K, Ito Y, Kojima M, et al. Intake frequency of fish and serum levels of long-chain n-fatty acids: a cross-sectional study within the Japan Collaborative Cohort Study. J Epidemiol. 2005;15:211. doi: 10.2188/jea.15.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wills-Karp M, Luyimbazi J, Xu X, et al. Interleukin-13: central mediator of allergic asthma. Science. 1998;282:2258. doi: 10.1126/science.282.5397.2258. [DOI] [PubMed] [Google Scholar]
  • 115.Wittamer V, Franssen JD, Vulcano M, et al. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J Exp Med. 2003;198:977. doi: 10.1084/jem.20030382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Woodruff PG, Boushey HA, Dolganov GM, et al. Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:15858. doi: 10.1073/pnas.0707413104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Woodruff PG, Modrek B, Choy DF, et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 2009;180:388. doi: 10.1164/rccm.200903-0392OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wu SH, Yin PL, Zhang YM, et al. Reversed changes of lipoxin A4 and leukotrienes in children with asthma in different severity degree. Pediatr Pulmonol. 2010;45:333. doi: 10.1002/ppul.21186. [DOI] [PubMed] [Google Scholar]
  • 119.Yang R, Fredman G, Krishnamoorthy S, et al. Decoding functional metabolomics with docosahexaenoyl ethanolamide (DHEA) identifies novel bioactive signals. Journal of Biological Chemistry. 2011;286:31532. doi: 10.1074/jbc.M111.237990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yokomizo T, Izumi T, Chang K, et al. A G-protein-coupled receptor for leukotriene B4 that mediates chemotaxis. Nature. 1997;387:620. doi: 10.1038/42506. [DOI] [PubMed] [Google Scholar]
  • 121.Zhen G, Park SW, Nguyenvu LT, et al. IL-13 and epidermal growth factor receptor have critical but distinct roles in epithelial cell mucin production. American Journal of Respiratory Cell & Molecular Biology. 2007;36:244. doi: 10.1165/rcmb.2006-0180OC. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES