Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 11.
Published in final edited form as: Nature. 2014 Jun 5;510(7503):92–101. doi: 10.1038/nature13479

Novel Pro-Resolving Lipid Mediators in Inflammation Are Leads for Resolution Physiology

Charles N Serhan 1
PMCID: PMC4263681  NIHMSID: NIHMS646633  PMID: 24899309

Preface

Advances on mechanisms in resolution of acute inflammation uncovered a new genus of pro-resolving lipid mediators that include separate families of molecules: lipoxins, resolvins, protectins and maresins, collectively coined specialized pro-resolving mediators (SPM). Synthetic SPM possess potent bioactions when administered in vivo. In animal experiments, SPM evoke anti-inflammatory and novel pro-resolving mechanisms as well as enhance microbial clearance. While identified in inflammation-resolution, SPM are conserved structures with functions also in host defense, pain, organ protection and tissue remodeling. This review covers SPM mechanisms and new omega-3 essential fatty acid pathways that open a path for physiologic functions.

Introduction

Today, excessive inflammation is widely appreciated as a unifying component in many chronic diseases including vascular diseases, metabolic syndrome, neurological diseases, and many others, and thus is a public health concern. Hence, understanding endogenous control points within the inflammatory response may give new views on disease pathogenesis and treatment approaches. Barrier break, trauma and microbial invasion each create the host’s need to neutralize invaders, clear the site, remodel and regenerate tissue. The acute inflammatory response is protective, a terrain where lipid mediators (LM) such as eicosanoids (prostaglandins (PG) and leukotrienes (LT)1)2 produced from the essential fatty acid arachidonic acid play critical roles in the initial response as do many cytokines and chemokines35. Interactions between prostaglandins, leukotrienes and pro-inflammatory cytokines amplify inflammation, where their pharmacologic inhibition and receptor antagonists reduce the signs of inflammation13. Yet, given that excessive inflammation contributes to many widely occurring diseases, improvements are needed.

Pathologists divide the acute inflammatory response into initiation and resolution (Fig. 1). Although resolution of disease is appreciated by clinicians, resolution itself was considered a passive process6. With identification of mediators with pro-resolving capacity biosynthesized from omega-3 essential fatty acids, evidence emerged that resolution of self-limited acute inflammation may be an active programmed response that is “turned on” in animal models and not simply a process of passive dilution of chemoattractants7,8. For a metabolite to fulfill the role of mediator it must be produced in sufficient amounts and in location(s) in vivo to evoke bioactions. The omega-3 fatty acids EPA and DHA, enriched in marine oils, have long been held to display anti-inflammatory properties where they compete with arachidonic acid reducing pro-inflammatory eicosanoids9. Their molecular mechanism(s) and whether omega-3 EPA and DHA are beneficial in human health and disease remains inconclusive. To this end, the possibility of utilization of omega-3 fatty acids (Fig. 1) by resolving inflammatory exudates to produce structurally distinct families of signaling molecules--namely resolvins, protectins and maresins, collectively termed SPM--opened a new interest in resolution pathways and innate immune mechanisms to attain homeostasis. SPM are agonists with the potential to stimulate key cellular events in resolution, namely cessation of PMN infiltration and enhance macrophage uptake of apoptotic cells5,6 in pre-clinical animal models10. This review addresses the role of novel lipid-derived SPM in resolution that also function in other systems, giving promise for SPM-pathways in resolution physiology toward human translation.

Figure 1. Lipid mediators in the acute inflammatory response, resolution and other outcomes.

Figure 1

LM play pivotal roles in the vascular response and leukocyte trafficking, from initiation to resolution. Eicosanoids are critical in initiating the cardinal signs of inflammation (upper left). The lipoxins, resolvins, protectins and maresins, specialized proresolving mediators (SPM), are produced in self-limited responses (Fig. 2). SPM stimulate cellular events that counter-regulate pro-inflammatory mediators and regulate PMN, monocyte and macrophage response, leading to resolution. Depicted are some pro-resolving actions in leukocyte trafficking (neutrophil-monocyte sequence), lipid mediator class switching and efferocytosis of apoptotic PMN that must occur in resolving exudates for restoration of normal structure and homeostasis. In addition to the release of n-3 substrate from phospholipid stores95, omega-3 substrates can enter mouse exudates via edema from peripheral blood57. SPM enhance efferocytosis, stimulate signs of resolution (lower right) and signal to adaptive immunity via lymphocytes. Failed resolution may lead to enhanced prostaglandins and leukotrienes, chronic inflammation and fibrosis. SPM counterregulate pro-inflammatory chemical mediators, reducing magnitude and duration of inflammation, and stimulate reepithelialization, wound healing, and tissue regeneration in model organisms.

Cellular Events in Resolution of Acute Inflammation

During the initiation phase, leukocytes traffic from the circulation, forming inflammatory exudates, traditionally viewed as battlefields6. First responders, neutrophils (PMN), swarm like bees to defend the host moving along chemotaxic gradients, e.g. leukotriene (LT)B411, exiting venules governed by prostaglandins (PGE2 and PGI2) acting on vascular cells and blood flow1 (Fig. 1). These lipid mediators, along with many cytokines, chemokines and complement components (C5a, C3b), stimulate chemotaxis of PMN into tissues to phagocytize and neutralize invaders36. Many current therapeutic agents block or antagonize initiation steps in acute inflammation (e.g. prostaglandin biosynthesis inhibitors or chemokine receptor antagonists)1,3,12. At the cellular level, the main events in resolution are cessation of PMN influx and macrophage clearance of debris including apoptotic neutrophils, a process known as efferocytosis4,5. Since the acute inflammatory response is protective, evolved to permit repair of injured tissues and eliminate invading organisms, it is ideally self-limited, leading to complete resolution enabling return to homeostasis (Fig. 1). Studying self-limited inflammation in animal models (inflammation that returns to homeostasis), and using a systems approach to interrogate resolving exudates, novel bioactive products were uncovered derived from essential fatty acids. These bioactions include limited PMN influx in vivo, reduce human PMN transmigration and counter-regulated TNFα and other cytokines in mice. Each was systematically evaluated, and the bioactive products are biosynthesized from EPA and DHA via separate pathways in human PMN and macrophages in vitro7,8,13.

Elucidation of Pro-Resolving Lipid-Derived Mediators: Pro-Resolving and Anti-Inflammatory Are Different

Anti-inflammation is not a process equivalent to proresolution, because proresolution involves SPM as agonists stopping further PMN influx and activation of nonphlogistic responses of macrophages and resolution programs (Fig. 1). Key to this paradigm change is identification of novel families of autacoids and their forms potentially triggered by aspirin, providing evidence that resolution is actively orchestrated by LM in animal models (Fig. 2 and Box 1). Challenges ahead are whether we can harness these pathways that stimulate resolution. The term resolvent and concept of promoting the disappearance of inflammation trace to the 10th century in the Canon of Medicine14,15. Also, because dietary n-3 supplements are widely used, with <25% directed by health care providers16, and clinical trials have given mixed results17, it is critical for public health to establish mechanisms that underlie their essential requirements.

Figure 2. SPM production in resolving inflammatory exudates.

Figure 2

Upper panel depicts a typical self-limited acute inflammatory response time course encountered in experimental settings from initiation (time 0) to resolution: edema, neutrophilic infiltration and nonphlogistic recruitment of monocytes/macrophages. Biosynthesis of SPM occurs temporally in resolving exudates. Non-phlogistic recruitment of monocytes and macrophages is required for homeostasis, repair and regeneration of injured tissues. Lower panel, Resolution n-3 metabolome. Biosynthesis of resolvins, protectins and maresins from EPA and DHA with the main bioactive structures from each family (see Box 1 and refs. 18,21 for details on biosynthetic mechanisms and stereochemical assignments of the bioactive products). Each SPM stimulates macrophage switching to M2 phenotype and is produced by human neutrophils, apoptotic PMN and macrophages13.

Box 1. Resolution n-3 metabolome.

The bioactive products were originally isolated from mouse resolving exudates, their structures elucidated and biosynthesis of each new n-3 family from EPA and DHA was recapitulated with isolated human neutrophils21 and macrophages in vitro23,24. Biosynthesis of E-series resolvins is initiated with molecular oxygen insertion at carbon-18 position of EPA produced by hypoxic human endothelial cells and acetylated COX-2 or P450. 18-HEPE is also produced by microbial P4507 (left), which is converted to bioactive E-series members by human neutrophils. Both aspirin-dependent and independent formation occurs in human peripheral blood47,61. Resolution metabolome also activates 17-lipoxygenation of DHA via hydrogen abstraction; 17S-HpDHA is converted to resolvin-epoxide-containing intermediates (center) that are transformed to resolvins D1-D4 by human PMN, each identified using physical methods including mass spectrometry of bioactive products. The 17-HpDHA intermediate is also precursor to 16,17-epoxide-protectin that is converted to protectin D1/neuroprotectin D1 and related structures by human leukocytes (PMN, T cells), neural cells and retinal-pigmented epithelial cells8,97,98,112. (Right): Maresins isolated from human and mouse macrophages are produced via initial lipoxygenation with molecular oxygen insertion at carbon-14 position to form the hydroperoxide intermediate that is rapidly converted to 13S,14S-epoxide-maresin and enzymatically converted to maresin 124. Lipoxygenase mechanisms involve hydrogen abstraction and molecular oxygen insertion at specific carbon positions that are predominantly in the S configuration. Aspirin, via COX-2 acetylation and P450 enzymes, contributes to the biosynthesis of R-configuration alcohols in lipoxins, resolvins and protectins37. The stereochemistry of each bioactive SPM family member shown is determined, and biosynthesis, potent pro-resolving and anti-inflammatory actions in murine exudates and human tissues confirmed (See ref. 18 for original reports and refs. 10,21).

Box 1

Using a systems approach with resolving mouse exudates was key to elucidating SPM actions and pathways4,8. Biosynthesis with human leukocytes and complete stereochemistry of each major resolvin (RvE1, RvD1, RvD2, RvD3 and RvD5) and protectin was accomplished by matching with material prepared by organic synthesis that also confirmed their potential bioactivity 10,18. Each controls the magnitude and duration of inflammation in vivo in animal disease models19, for example increasing survival (Fig. 2). The potent in vivo actions of RvD1 and RvD2 are reported in many organ pathologies and processes including vascular20, airway21, dermal, renal, ocular, pain, obesity, fibrosis and wound healing10,22. Governing PMN influx, resolution macrophages and reducing pro-inflammatory mediators (i.e. PAF, LTB4, PG) appear fundamental in all organs.

Newest Resolvents: Resolvin D3 and Maresin Pathway

Within self-limited exudates, RvD3 displays a unique timeframe compared to RvD1 and RvD2. RvD3 appears late in resolution in mouse peritonitis, suggesting a specific role. RvD3’s complete stereochemistry was recently established14, confirming its potent anti-inflammatory and proresolving actions8. Macrophage biosynthesis of MaR1 and its potent proresolving and tissue regenerative actions23 (Fig. 2) involve a 13S,14S-epoxide-maresin intermediate that is also active, and stimulating M1 to M2 phenotype-switch (Fig. 2)24. The switch in macrophage phenotype toward M2 is associated with reparative and anti-inflammatory MΦ functions5,25.

Resolution Agonists and Resolution Disruptors

Several lessons emerge from recent studies. PG are central to vascular responses, permitting PMN and monocytes to leave postcapillary venules, diapedesis. Their production via COX-1 and COX-2 is critical for initiation and timely resolution (Fig. 1)26,27. PGE2 and PGD2 each evoke pro-inflammatory and anti-inflammatory responses that depend on location12. PGE2 enhances LTB4-mediated PMN extravasation and tissue injury that is blocked, for example, by topical administration of synthetic lipoxin A4 (LXA4) and its aspirin-triggered epimer 15-epi-LXA428, illustrating a pro-inflammatory PGE2 function in mouse skin and ability of 15-epi-LXA4 mimetics to limit PMN infiltration and tissue injury. LC-MS-MS-based profiling demonstrated the temporal switch from PG and LTB4 to appearance of lipoxins, a process coined lipid-mediator- (LM)-class switching, within mouse exudates (Fig. 1, Box 1). PGE2 or PGD2 added to isolated human PMN increase 15-lipoxygenase type I translation from mRNA stores in a cAMP-dependent manner, increasing LX biosynthesis identified by MS-MS spectra26.

Inhibition of COX-2 delays resolution because prostaglandins play critical roles in resolution and are also initiators of LM-class switching (Fig. 1) in animal disease models in vivo19,26,29. In mapping resolution, it became apparent that initiation signals the end of inflammation (alpha signals omega)4 and that leukocyte traffic in pus permits prostanoids to signal biosynthesis of other resolution mediators (Figure 1 & 2). For example, disruption of physiologic LM-class switching has deleterious consequences in mouse arthritis29.

To pinpoint critical steps and mechanisms of SPM action within inflammation-resolution, it was important to introduce quantitative indices19,30 that enable assessment of resolution in vivo21,27,31. Resolution indices identified agents that stimulate as well as those that disrupt or delay resolution (resolution interval, Ri), e.g. COX-2 and lipoxygenase inhibitors19,27,32. Specific SPM shorten Ri by limiting PMN recruitment and stimulating both macrophage efferocytosis (Fig. 1) and bacterial killing31,33,34, demonstrating the PMN-monocyte sequence and macrophage responses needed for tissue regeneration23. Glucocorticoids, specific cyclin-dependent kinase inhibitors, statins, annexin peptides and aspirin, enable resolution31,35,36. As there are many mediators in the initiation of inflammation, there are many endogenous mediators and drugs that impact resolution5,19,21.

Aspirin and NSAIDs inhibit prostanoid biosynthesis, but aspirin is an irreversible inhibitor that acetylates COX, while NSAIDs are reversible inhibitors1,2. Aspirin acetylation of COX-2 modifies the catalytic domain, blocking PG-biosynthesis, which is well known1,2, yet remains active producing 15R-HETE from arachidonic acid, 18R-HEPE from EPA and 17R-HDHA from DHA in cells carrying COX-2. These can be transformed by human neutrophils in vitro to aspirin-triggered lipoxins, aspirin-triggered resolvins7,8 and aspirin-triggered protectins37. Each potently stops human PMN migration and enhances macrophage cleanup, enhancing resolution in mice. Whether aspirin or statins enhance production of aspirin-triggered-SPM in humans remains to be established using mass spectral-based identification. Also, whether aspirin favors resolution in humans, where distinct resolution-phenotypes emerged, is of considerable interest27,38. In mice, intravascular LXA4 is produced via platelet-neutrophil aggregates during ischemia, which reduces vascular inflammation. Aspirin triggers 15-epi-LXA4 identified using LC-MS-MS, which was less effective in ALX receptor-deficient mice, providing in vivo evidence that aspirin can jumpstart resolution circuits in mice39.

New Mechanisms in Local SPM Biosynthesis

Microparticles (MP) are membrane-derived vesicles produced by a range of cell types that contribute to human pathologies. MP from self-resolving exudates display anti-inflammatory and proresolving capacity40 in mice. Resolution-MP enhance efferocytosis13,40 and carry pro-resolving signals including hydroxy-SPM-intermediates esterified in phospholipids 40. Secretory PLA2 release these from MP for transcellular conversion by human macrophages in vitro13,40. Since nanomedicines are of interest, resolution-MP and their ability to shorten Ri in mouse peritonitis were used as a basis for biomimicry to construct humanized nanoparticle-containing LXA4 analog or AT-RvD140. These nano-proresolving medicines (NPRM) carrying SPM or SPM-analogs, enhance wound healing of human keratinocytes and are protective in a mouse model of temporomandibular joint disease characterized by inflammation-induced bone loss40 (Figure 1, Box 2).

Box 2. Key Evidence for n-3 Pro-Resolving Mediator Structure-Function and Steps Toward Human Translation.

Outline of the evidence and systems used to identify the new n-3-derived molecules and their bioactions from resolving inflammatory exudates in mice, structure elucidation and actions with human PMN, and proposed biosynthesis from EPA and DHA using 18Oxygen, 14Carbon and trapping of transient intermediates18. Confirmation of the bioactive structures and total organic synthesis to assign complete stereochemistry of each SPM family member required developing a strategy to match the biologically active isolated product with those prepared by total organic synthesis, where NMR confirmed the geometry of the conjugated double bond systems present in each, e.g. RvE147, RvD1, RvD234 or MaR123,24 (see ref. 18 for matching criteria and Illustration for Box 2). SPM resolving functions are separate from anti-inflammatory; to serve as a SPM, the n-3 product must be biosynthesized at the in vivo levels commensurate with their bioactions (inset, left). For many of the SPM, this criterion has been achieved and confirmed with commercial resolvins (see ref. 10).

SPM are conserved structures present in trout (PD1 and RvD1)88, salmon (RvD1 and RvD2)117 and planaria23. Microfluidic chambers that permit visualization of cell-cell interactions between leukocyte subpopulations (i.e. human PMN and monocytes) and distinguish phlogistic vs. nonphlogistic phagocyte behavior are ideal to screen SPM and humanized NPRM40,56. Single cell screening with microfluidic devices permits optimization for enriching MP with SPM and production of NPRM as well as viewing neutrophil-monocyte interactions56,57 essential for appreciating signals for the PMN-monocyte sequence Figure 1.

With microfluidic chambers, single human PMN were assayed within ~5 mins of their capture from whole blood (minimizing isolation time reduces potential artifacts). Each SPM, in nanomolar concentrations, stops human PMN migration to IL-8, where at equimolar doses the precursor (i.e. DHA) is not active56,57. Resolution indices permit quantitative assessment of the actions of SPM in animal models19,30 that is essential to defining SPM within the integrated response of the host to acute inflammatory challenges. Demonstration and identification of n-3 SPM in human tissues is a required step to appreciate their potential roles in humans. RvE1 and RvE247,61 in peripheral blood of healthy volunteers, with some given EPA supplements, were identified using LC-MS-MS multi-reaction monitoring. RvD1 and RvD2111, PD1 and 17-HDHA were identified in human exhaled breath condensates from asthmatics43 and additional SPM identified from human adipose118, Alzheimer’s disease brain96,101, multiple sclerosis patients119 and rheumatoid arthritis120 using mass spectral identification. While the demonstration of n-3 in human tissues is at present at the level of first-time identification(s), with the capabilities of LC-MS-MS-based profiling13, SPM also have potential as markers for nutritional status. The field is now set with tools for assessing SPM function in humans and their relationship(s) to nutrition and human disease.

MP can also transfer substrate and intermediates to macrophages (MΦ) during efferocytosis enhancing SPM biosynthesis, demonstrated by transfer of deuterium label from precursors to labeled SPM identified using LC-MS-MS13. Myeloid cells at different stages display agonist- and phenotype-specific LM profiles. For example, human PMN from healthy peripheral blood produce predominantly LTB4, while apoptotic PMN produce PGE2, LXB4 and RvE2 signals.13

Both M1 and M2 macrophages display specific markers and pathways specialized to their functions of MΦ subpopulation in inflammation and resolution25. Human M2 macrophages possess increased enzymes41 needed for cell type-specific LM. M2 produce SPM with lower LTB4 and PG than M1. Both cell types engulf apoptotic PMN, changing their LM. In M2, LTB4 is down-regulated and SPM increased13, suggesting M1 and M2 subpopulation25,41 produce functional LM signatures that can impact both physiologic and pathophysiologic states13. Also, secreted PLA2 group IID was identified as a resolving sPLA2 expressed in dendritic cells and macrophages that releases substrates with the capacity for producing RvD1 and PGJ2 identified by mass spectrometry in mouse lymphoid tissue in vivo. 42 The functional contributions of this resolving sPLA2 to the inflammatory response in humans remains of interest.

Eosinophils are well appreciated in parasitic infections and allergic responses. In severe asthma, PD1 is present in human exhaled breath condensates43 and is decreased in human eosinophils from patients with severe asthma44. Human eosinophils produce PD1, which reduces adhesion molecules (CD11b and L-selectin), eotaxin-1/CCL11 and chemotaxis at nanomolar concentrations, without affecting degranulation, superoxide generation or cell survival. Eosinophils also stimulate resolution in mouse peritonitis via SPM initiated by mouse eosinophils45. LC-MS-MS-lipidomics identified LXA4, RvD5, 17-HDHA and PD1 from eosinophils and RvE3 in vivo (Fig. 2) that limit PMN infiltration and regulate MΦ4446. Hence, via their ability to produce SPM, eosinophils may contribute to resolution. To support this, Arita and colleagues ref 47 found eosinophil depletion leads to deficit resolution rescued by PD1 or eosinophil restoration in mice. Thus, cellular traffic to inflammatory loci has a dynamic impact on LM signatures and specific SPM metabolomes activated within local milieu.

SPM Cellular Actions in Disease Models

SPM increase survival in diverse mouse models. In murine models, airway, dermatologic, ocular, organ-specific inflammation and tissue injury resulting from collateral damage are improved with exogenous SPM10. The SPM nanomolar-doses required to stop ongoing inflammation and promote resolution rely on GPCR. Several SPM receptors are identified using GPCR screening, labeled-ligands for specific binding (stereospecific nM Kd) and functional cellular responses 4749. SPM in general do not utilize intracellular Ca2+ mobilization in PMN for signal transduction but instead activate phosphorylation. RvE1 specifically binds to ChemR2347 and BLT1 to evoke pro-resolving responses. RvE1 activation of ChemR23 enhances macrophage phagocytosis via phosphoprotein-mediated signaling48. RvE1 blocks LTB4 binding and also signals via BLT1 to promote apoptosis of PMN for their clearance by MΦ49, while LTB4-BLT1 signals PMN survival. PMN RvE1 signaling involves blocking survival signals, an important difference for PMN in the innate response, where they must undergo timely apoptosis for clearance5,49 (Fig. 2).

RvD1 binds and activates human GPR32 and shares human and murine LXA4 receptor (ALX/FPR2). Transgenic mice overexpressing human ALX-FPR2 require less RvD1 to stop inflammation50, and in receptor-deficient mice, RvD1 is apparently without leukocyte-directed actions51. Resolution involves specific miR, regulated by SPM receptors50,52,53. RvD1-GPR32 upregulates miR-208 and anti-inflammatory IL-10 as well as down-regulates miR-219, decreasing LTB4 via regulation of 5-lipoxygenase52. miR regulation by SPM is an example of SPM-signaling that can have a sustained tissue impact.

SPM-receptors rapidly signal as well. For example, recombinant RvD1-GPR32 blocks histamine receptor (H1)-stimulated increases in intracellular Ca2+ in CHO cells via rapid stimulation of phosphorylation of H1 receptor, stopping Ca2+ mobilization54. This form of SPM signaling, documented with conjunctival goblet cells and RvD1, is also functional in salivary glands55 and likely to be relevant in human PMN, which rapidly stop chemotaxis in microfluidic chambers and change shape on exposure to SPM56,57. RvD3 and RvD5 can also activate recombinant GPR3214,33. Given the temporal production of RvD3 in vivo14 (Fig. 2), these findings underscore that SPM produced locally can impact different cell types and receptors in a spatial-temporal dependency.

In addition to RvD1 and LXA4, ALX/FPR2 is also activated by peptide pro-resolving mediators, e.g. annexin A1, as well as pro-inflammatory peptides, at higher concentrations36. This capacity of ALX/FPR2 involves ligand-biased receptor activation with heterodimerization of ALX with related FPR dictating pro-inflammatory signaling, and ALX homodimer gives pro-resolving signaling36. LXA4 also enhances ALX/FPR2 promoter activity, which has a mutation in human cardiovascular disease58.

Infection and Resolution Programs

Topical RvE1 and LXA4 each reduce severity of periodontal disease in rabbits by enhancing P. gingivalis clearance, causative organism in this infection59,60. While anti-inflammatory actions of SPM were uncovered in sterile-inflammation models7,8, the relation between resolution and infection is of interest because of the known eventual immunosuppressive actions of anti-inflammatory drugs12. Surprisingly, RvD2 protects mice from cecal ligation-puncture (CLP)-induced sepsis34, with potent actions enhancing phagocytosis and bacterial killing. In self-limited E. coli infections, resolution programs are activated in mice and host PD1, RvD5 and RvD1 are elevated33. When added back to mouse phagocytes, human MΦ or PMN, SPM enhance bacterial phagocytosis and killing as well as clearance33,34,61. Importantly, SPM, acting on the host, lower antibiotic doses needed to clear infections.

Exogenous LXA4 is also protective in rat CLP, reducing bacterial burden and pro-inflammatory mediators via a MΦ NFκB-mediated mechanism reducing systemic inflammation62. Aspirin-triggered-LXA4 increases E. coli phagocytosis in a PI3K- and scavenger receptor-dependent manner, and ALX/FPR2 is upregulated in patients with Crohn’s disease and enhances bacterial clearance63. Mycobacterium tuberculosis infections are susceptible to modulation of leukotriene A4 hydrolase. Besides altering LTB4, this may also engage resolution programs via activating LTB4-LXA4 production, regulating host responses in zebrafish, mice and possibly in humans64,65. Given the importance of rising antibiotic resistance, activation of resolution programs could provide new anti-microbial approaches to lower antibiotic exposure33.

Herpes simplex virus causes ocular infections that lead to stromal keratitis with viral-initiated immunopathology. RvE1 and PD1 are each potent and topically active in this infectious mouse model, reducing pro-inflammatory mediators and stimulating IL-1066,67. H5N1 virus lethal dissemination activates genes in mice tracked to LX biosynthesis, where sustained inflammation inhibits LX-mediated anti-inflammatory host responses, permitting viral dissemination68. H5N1 activates host resolution-metabolome increasing PD1, identified by LC-MS-MS69. Host protectins display antiviral activity blocking replication of H5N1 influenza virus. During the time course of H3N2, a low-pathogenicity strain of influenza, anti-inflammatory mediators are produced with infection that correlates with resolution and SPM-related pathway-markers70. SPM are also found in yeast infections, e.g. Candida, where RvE1 enhances yeast killing and clearance in mice71. These results enforce the notion that treating the host during infection with host-directed pro-resolving molecules could open new opportunities in host-pathogen interactions to reduce inflammation and enhance clearance33,61,72.

Chronic Inflammatory Disease Models

Periodontal disease is a chronic inflammatory disease, where infection leads to PMN-mediated tissue injury around the tooth. Activated PMN from periodontitis patients produce PGE2, LTB4 and LXA4, each identified using LC-MS-MS73. PGE2 in this tissue leads to bone loss. P. gingivalis elicits PMN recruitment in mouse air pouches along with COX-2 upregulation. LXA4 stable analogs reduced both PMN influx and COX-2 expression with the oral pathogen73. Also, P. gingivalis increased COX-2 expression in mouse lung and heart, and 16S ribosomal RNA of P. gingivalis was present in these tissues, supporting a role for this oral pathogen in development of systemic inflammation. Transgenic (TG)-rabbits overexpressing human 15-lipoxygenase type I produce 6–10 times the amounts of LXA4 identified using LC-MS-MS than non-TG-rabbits59; TG-rabbits show less bone loss in periodontitis and markedly reduced PMN recruitment and vascular leakage through their skin on challenge, suggesting overexpression of LX biosynthesis is protective and may be useful in controlling inflammation-mediated bone destruction59.

Unexpectedly, overexpression of 15-lipoxygenase in these transgenic-rabbits sharply reduces atherosclerotic lesions74. In 12/15-lipoxygenase tg-mice, LC-MS-MS identified RvD1, PD1 and 17-HDHA with reduced PGE2 from activated macrophages75. LXA4, PD1 and RvD1 each reduced cytokines (e.g. MCP-1) from endothelial cells and adhesion molecules (P-selectin, VCAM-1) but not ICAM-1. They also enhanced uptake of apoptotic thymocytes, which could contribute to the anti-atherogenic role of this pathway in mouse. This process can also be subject to dietary manipulation to govern severity of atherogenic lesions75,76. RvE1 is protective in periodontal disease but, unlike LXA4, exogenous RvE1 also stimulates bone regeneration in rabbit periodontitis60,77. In murine arthritis, RvD1 and 17-HDHA reduce pain and tissue damage, proving more potent than either steroid or anti-pain treatments78.

Unresolved inflammation, epithelial and microvascular injury can lead to excessive fibrosis (Fig. 1) that impairs organ function. Leukotrienes are profibrotic and in humans with scleroderma interstitial lung disease, LXA4 is in bronchoalveolar lavages at levels that appear unable to counter-regulate profibrotic factors79. In animals, exogenous aspirin-triggered-LX analog reduces bleomycin-induced pulmonary fibrosis80, and both LXA4 and benzo-LXA4 reduce renal fibrosis81. Exogenous RvE1 and RvD1 protect from renal fibrosis, reducing collagen I and IV, α-SMA and fibronectin82. Also, exogenous RvD1 reduces pro-inflammatory mediators that are generated in response to cigarette smoke and pulmonary toxicants.83

Organ Regeneration and Wound Healing

Exogenous LXA4 stimulates reepithelialization of cornea in a gender-specific fashion in mice84. Exogenous RvE1, RvD1 and RvD2 each stimulate dermal wound healing, reducing neutrophilic infiltration and stimulate reepithelialization of murine skin wounds when applied to wounds85. Exogenous RvD1 and RvD2 also stimulate diabetic wound healing in mice22,86. Given the role of macrophages in wound healing and organ regeneration, the macrophage-derived maresin pathway stimulates tissue regeneration. The maresin pathway (Figure 2) is present in planaria Dugesia tigrina, a Platyheminthes used in regeneration studies. RvE1 and MaR1 each reduce regeneration times (speed of regrowing head segments)23. Given the importance of tissue regeneration in trauma and infection, regulation of resolution programs may hold promise.

SPM in Adaptive Immunity

Lymphoid tissue, e.g. mouse spleen, produces RvD1, 17-HDHA, PD187 and LXA488 from endogenous sources identified using mass spectrometry, suggesting they’re strategically positioned to act on lymphocytes (Fig. 1). Both exogenous 17-HDHA and RvD1 increase human B cell IgM and IgG, a response not shared by PD1. Exogenous 17-HDHA augments B cell differentiation toward CD27(+)CD38(+) antibody-secreting cell phenotype87. PD1 is biosynthesized by human T helper 2-skewed mononuclear cells via 16(17)-epoxy-protectin intermediate (Fig. 2) and reduces T cell migration, TNFα and INFγ, promoting T cell apoptosis89. Exogenous LXA4, RvE1 and PD1 each upregulate CCR5 expression on leukocytes that bind chemokines, facilitating their clearance and resolution90. Exogenous PD1 reduces CD4+ T cell infiltration into cornea66, as does RvE1 in Herpes simplex viral infections67. Exogenous RvD1 reduces CD11b+ leukocytes and CD4+ and CD8+ T lymphocytes within the eye in uveitis91. Exogenous RvE1 and RvD1 each regulate T-cell activation in choroid-retina and are biosynthesized in this tissue identified by LC-MS-MS92. Exogenous RvE1 induces apoptosis of activated T cells via 2,3-dioxygenase induction in DC giving a new functional DC-subtype in resolution93. Exogenous RvE1 reduces mouse CD4+ T cells and CD8+ T cells in atopic dermatitis94.

Neuroinflammation and Pain

Mouse and human brain have the capacity to produce resolvins and protectins, as do human micro glial cells where they may reduce cytokine expression8,95,96. Their production by trout brain cells indicates SPM are conserved from fish to humans88. In mouse ischemic stroke, immunoreactive resolvins, protectins and their aspirin-triggered forms are produced97, where synthetic compounds are protective, down regulating excess leukocyte infiltration, and reduce local neuronal injury, COX-2 induction, IL-1β and NFκB. Thus, in brain, DHA is a potential precursor to neuroprotective signaling pathways evoked by ischemia-reflow tissue injury. Given its potent actions to reduce neuroinflammation and protect neural cells, this 10,17-dihydroxy-protectin (a docosatriene) was coined neuroprotectin D1 with Bazan and colleagues when biosynthesized and acting in neural tissues and retinal epithelial cells98.

DHA is enriched in brain, synapses and retina, where its protective role is appreciated, yet its role as a precursor to mediators in resolution and neuroprotection is still emerging. Bazan and colleagues showed potent protective roles of synthetic NPD1 in the nervous system, reducing stress pathways that lead to cell death and increase cell survival, and in several ocular models of important diseases NPD1 targets microglia8,98,99. Human PMN biosynthesize 17R-NPD1/PD1 that is enhanced by aspirin in vitro, which limits PMN transmigration, and enhanced MΦ efferocytosis37. Synthetic -17R-NPD1 reduces brain edema in the penumbra and subcortical lesion size and improves neurological scores100.

In human Alzheimer’s disease (AD), brain NPD1 is reduced96. Also, resolution-pathway (SPM-receptors and products) are diminished in brain from AD101. LXA4 and RvD1 are reduced in cerebrospinal fluid and hippocampus that correlated to mini-mental state examinations in these patients. These findings provide further evidence that failed resolution may contribute to human disease101. RvD1 added to MΦ from AD patients in vitro reduces their pro-inflammatory phenotype and enhances phagocytosis of amyloid-beta102, consistent with the possibility that resolvins promote clearance of amyloid-beta deposition to reduce inflammation in AD. Hence, SPM may play homeostatic roles in brain and peripheral tissues, each with selective functions to reduce neuroinflammation.

Inflammation can evoke pain that may persist. Synthetic lipoxins reduce pain in murine models, LXA4 receptor (ALX/FPR2) is on spinal astrocytes, and local spinal LXA4, LXB4 or their metabolically stable analogs reduces inflammation-induced pain103. Each SPM dampens pain, having specific targets of action104 in mice, demonstrated with RvE1 and RvD1 for inflammatory pain involving both central and peripheral sites105. RvE1 administered intrathecally in mice is more potently analgesic than morphine or COX-2 inhibitor. RvE1 receptor (ChemR23) is present in DRG, where synthetic RvE1 regulates pERK-dependent TRPV1inhibition and TNFα-mediated hyperalgesia centrally. In postsynaptic neurons, synthetic RvE1 inhibits glutamate and TNFα stimulation of NMDA-R and mechanical allodynia105. Synthetic RvD1, RvD2 and PD1/NPD1 each reduce pain via inhibition of specific TRPV-channels106,107.

MaR1 inhibits TRPV1 in neurons and blocks capsaicin-induced inward current (IC50 0.49 nM), diminishing inflammatory and chemotherapy-evoked neuropathic pain in mice24. Both AT-RvD1 and 17R-HDHA reduce adjuvant-induced arthritis in rats and associated pain78, reducing NFκB and COX-2 expression in spinal cord, and within arthritic joints reduce TNFα and IL-1β. In addition to leukocytes and microglia, SPM receptors are present on neuronal bodies, nerve terminals (skin and muscle) and synaptic terminals, where they regulate specific TRP channels. For example, RvE1-ChemR23 interaction in DRG regulates TRPV1, but not via direct activation of channels like endocannabinoids104 or other lipids that act to directly bind TRP-channels; rather each SPM activates specific GPCR in pico-nanomolar range to regulate channels involved in pain signaling.

Direct comparisons between synthetic LXA4 and AT-RvD1 in rat mechanical hypersensitivity in inflammation-induced pain indicate that both effectively reduce hypersensitivity and pro-inflammatory mediators from astrocytes108. Cognitive decline following major surgery or critical illness is a major public health concern. Cognitive decline results from local increases in pro-inflammatory mediators. Systemic AT-RvD1 prophylaxis improves memory decline in a mouse surgery model, protecting from postoperative neuronal dysfunction109. Whether SPM actions in mouse pain models translate to reducing human pain and improve cognition remains of interest.

Towards human translation

Since resolvins and protectins were identified in mouse exudates, it was essential to establish their biosynthesis by human leukocytes and in human tissues7,8,95 (Box 2). RvE1 and RvE2 are identified using mass spectrometry in human blood47,61,110, providing substrate is available. RvD1 and RvD2 were determined in human plasma and serum 111. The capacity to form Resolvins (RvD1, RvD2) and protectins (PD1 and 10S,17S-diHDHA112, a.k.a. PDx) in murine placenta was confirmed by mass spectrometry and was increased by dietary omega-3113. Another strategic location for SPM is in human breast milk114, where LXA4, RvD1 and RvE1 were identified using mass spectrometry in milk from mothers during the first month of lactation114. These identifications, made possible with LC-MS-MS and availability of SPM, open opportunities for rigorous assessment of their functional roles and potential in human physiology.

Recently, Markworth et al.115 provided evidence for LM-class switching in humans. Using a strenuous resistance exercise protocol, venous blood was collected during the time course post exercise and lipid mediators present in peripheral blood were identified using LC-MS-MS-lipidomics. Initial post-exercise recovery phase demonstrated the presence of prostanoids temporally followed by leukotrienes and p450-derived eicosanoids (EETs), as well as lipoxins, resolvins and protectins. Widely used for muscle aches and pains, the NSAID ibuprofen blocked exercise-induced prostanoids as well as reduced LTB4 and both delayed and diminished appearance of SPM identified by mass spectrometry. Resistance-exercise in humans illustrates the acute pro-inflammatory mediators, presumably from muscle, and their potential link to resolution programs115. However, resolvins and protectins were reported in relative amounts, and additional evidence is needed to establish their levels in healthy exercising individuals.

In a 60-patient double-blind trial116 with infantile eczema, topical 15(R/S)-methyl-LXA4 relieved severity and improved quality of life without apparent adverse events. In these infants, lipoxin-analog was as effective as topical steroid.

Looking Forward

It’s when and where that counts for autacoids, and it’s important to emphasize that the first response in acute inflammation is ubiquitous and mounts throughout the body. SPM are agonists of resolution. Each stimulates cessation of PMN influx, efferocytosis and enhances phagocytosis for microbial containment: signs of resolution (Fig. 1). These are the defining SPM functions. Each SPM-pathway possesses additional nonredundant functions on target cell types. At the cellular and molecular level, SPM counterregulate pro-inflammatory mediators (eicosanoids, chemokines, cytokines33 and adipokines22); regulate specific miR53, cell traffic and enhance microbial killing via receptor-mediated mechanisms in animal models in vivo and with human neutrophils and macrophages10,13,22.

Results from pre-clinical disease models10 suggest treatment of inflammation-associated disease may be possible with SPM-agonists that stimulate resolution and protect organs from collateral damage (Box 2). RvE1, MaR1 and NPD1/PD1 are each in clinical development programs licensed by this institution. RvE1 mimetic is in human Phase III for ocular indications1, and NPD1/PD1 for neurodegenerative diseases2, given their ability to regulate inflammation-resolution without immunosuppression.

Since the means to identify SPM in human tissues uses LC-MS-MS-based approaches and internal standards that only recently became available, at this point relatively few studies demonstrate SPM in human tissues (e.g. blood, milk, adipose tissue and brain). Hence, evidence for SPM formation in humans is very early stage and their functional importance in human health and diseases remain to be established. Whether SPM have physiologic actions in target tissues in humans, given that they are produced in levels that display potent selective actions in animals10,20, can now be addressed (Box 2) using LC-MS-MS-based LM-SPM profiling of human tissues. Also, the impact of antiinflammatories in these pathways can be addressed in humans with this approach. Given that human neutrophils undergoing apoptosis and macrophages produce SPM identified by LC-MS-MS profiling13 without n-3 EFA supplementation, it will now be possible, with the sensitivity of LC-MS-MS, to determine individual human SPM profiles (personalized LM-SPM-metabolomics). To advance this field, it is now possible to assess SPM production in humans using LC-MS-MS-based LM-SPM translational metabolomics7,13 and determine their causal relationship(s) to host-resolution mechanisms. It is also important to determine, in healthy individuals, n-3 EFA supplementation and doses that may increase or diminish SPM within specific tissues. Importantly, it is critical to assess whether human disease(s) characterized by excessive inflammation results from failed resolution mechanisms via the absence of specific SPM-pathways and whether these can be rescued either by substrates or via therapeutic versions of SPM mimetics. Given that resolution of inflammation is fundamental for all organs, new approaches to stimulating resolution with potential use of SPM as pathway-markers are now also approachable, as well as determining SPM relationship(s) to nutrition in humans.

SPM emerge from animal models as potential regulators in physiologic pathways in resolution and unresolved inflammation that can impact infection, pain, obesity, organ protection22 and inflammatory diseases (Box 2) beyond the roles of their n-3 EFA precursors in intermediary metabolism and membrane dynamics. Identification of SPM bioactive-metabolomes and appreciation that exudates drive resolution in part via SPM sets a new terrain to evaluate resolution physiology and pharmacology, where SPM are vital as chemical signals for catabasis and host defense.

Acknowledgments

The author thanks Drs. Sarah Orr, Jesmond Dalli and Nan Chiang for critical reading of this manuscript and grants from the NIH (R01GM038765 and P01GM095467) and the Mérieux Foundation (France) for support of the author’s research. I also thank our collaborators and investigators contributing to this area whose publications were not cited due to size limitations.

Footnotes

1

Auven Therapeutics. RX 10045 – ocular inflammation. http://www.auventx.com/auven/products/rx10045.php

2

Anida Pharma Inc. Neuroprotectin D1. http://www.anidapharma.com/lead-molecule.html

Competing financial interests: CNS is an inventor on patents [resolvins] 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.

References

  • 1.Flower RJ. Prostaglandins, bioassay and inflammation. Br. J. Pharmacol. 2006;147:S182–S192. doi: 10.1038/sj.bjp.0706506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Samuelsson B. Role of basic science in the development of new medicines: examples from the eicosanoid field. J. Biol. Chem. 2012;287:10070–10080. doi: 10.1074/jbc.X112.351437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dinarello CA, Simon A, van der Meer JW. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nature reviews. Drug discovery. 2012;11:633–652. doi: 10.1038/nrd3800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.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]
  • 5.Maderna P, Godson C. Lipoxins: resolutionary road. Br. J. Pharmacol. 2009;158:947–959. doi: 10.1111/j.1476-5381.2009.00386.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cotran RS, Kumar V, Collins T, editors. Robbins Pathologic Basis of Disease. W.B. Saunders Co; 1999. [Google Scholar]
  • 7.Serhan CN, 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. J. Exp. Med. 2000;192:1197–1204. doi: 10.1084/jem.192.8.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Serhan CN, et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter pro-inflammation signals. J. Exp. Med. 2002;196:1025–1037. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lands WEM. Fish, Omega-3 and Human Health. 2nd edn. AOCS Press; 2005. [Google Scholar]
  • 10.Serhan CN, Chiang N. Resolution phase lipid mediators of inflammation: agonists of resolution. Curr. Opin. Pharmacol. 2013;13:632–640. doi: 10.1016/j.coph.2013.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Malawista SE, de Boisfleury Chevance A, van Damme J, Serhan CN. Tonic inhibition of chemotaxis in human plasma. Proc Natl Acad Sci U S A. 2008;105:17949–17954. doi: 10.1073/pnas.0802572105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fullerton JN, O'Brien AJ, Gilroy DW. Lipid mediators in immune dysfunction after severe inflammation. Trends in immunology. 2014;35:12–21. doi: 10.1016/j.it.2013.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dalli J, Serhan CN. Specific lipid mediator signatures of human phagocytes: microparticles stimulate macrophage efferocytosis and pro-resolving mediators. Blood. 2012;120:e60–e72. doi: 10.1182/blood-2012-04-423525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Dalli J, et al. Resolvin D3 and aspirin-triggered resolvin D3 are potent immunoresolvents. Chem. Biol. 2013;20:188–201. doi: 10.1016/j.chembiol.2012.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Serhan CN. The resolution of inflammation: the devil in the flask and in the details. FASEB J. 2011;25:1441–1448. doi: 10.1096/fj.11-0502ufm. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bailey RL, Gahche JJ, Miller PE, Thomas PR, Dwyer JT. Why US Adults Use Dietary Supplements. JAMA Intern Med. 2013;173:355–361. doi: 10.1001/jamainternmed.2013.2299. [DOI] [PubMed] [Google Scholar]
  • 17. Yates CM, Calder PC, Ed Rainger G. Pharmacology and therapeutics of omega-3 polyunsaturated fatty acids in chronic inflammatory disease. Pharmacol. Ther. 2013 doi: 10.1016/j.pharmthera.2013.10.010. A thoughtful review of the state-of-the-art of omega-3 essential fatty acids in inflammatory diseases and their potential in supplementation, dietary manipulation, and pharmacologic uses.
  • 18.Serhan CN, Petasis NA. Resolvins and protectins in inflammation-resolution. Chem. Rev. 2011;111:5922–5943. doi: 10.1021/cr100396c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.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]
  • 20.Miyahara T, et al. D-series resolvins attenuate vascular smooth muscle cell activation and neointimal hyperplasia following vascular injury. FASEB J. 2013;27:2220–2232. doi: 10.1096/fj.12-225615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Levy BD, Serhan CN. Resolution of acute inflammation in the lung. Annu. Rev. Physiol. 2014;76:27.21–27.26. doi: 10.1146/annurev-physiol-021113-170408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Spite M, Claria J, Serhan CN. Resolvins, Specialized Proresolving Lipid Mediators, and Their Potential Roles in Metabolic Diseases. Cell metabolism. 2013 doi: 10.1016/j.cmet.2013.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Serhan CN, et al. Macrophage pro-resolving mediator maresin 1 stimulates tissue regeneration and controls pain. FASEB J. 2012;26:1755–1765. doi: 10.1096/fj.11-201442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dalli J, et al. The novel 13S,14S-epoxy-maresin is converted by human macrophages to maresin1 (MaR1), inhibits leukotriene A4 hydrolase (LTA4H), and shifts macrophage phenotype. FASEB J. 2013;27:2573–2583. doi: 10.1096/fj.13-227728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–896. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
  • 26.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]
  • 27.Morris T, et al. Effects of low-dose aspirin on acute inflammatory responses in humans. J. Immunol. 2009;183:2089–2096. doi: 10.4049/jimmunol.0900477. [DOI] [PubMed] [Google Scholar]
  • 28.Takano T, Clish CB, Gronert K, Petasis N, Serhan CN. Neutrophil-mediated changes in vascular permeability are inhibited by topical application of aspirin-triggered 15-epi-lipoxin A4 and novel lipoxin B4 stable analogues. J. Clin. Invest. 1998;101:819–826. doi: 10.1172/JCI1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chan MM-Y, Moore AR. Resolution of inflammation in murine autoimmune arthritis is disrupted by cyclooxygenase-2 inhibition and restored by prostaglandin E2-mediated lipoxin A4 production. J. Immunol. 2010;184:6418–6426. doi: 10.4049/jimmunol.0903816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bannenberg GL, et al. 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]
  • 31.Lucas CD, et al. Downregulation of Mcl-1 has anti-inflammatory pro-resolution effects and enhances bacterial clearance from the lung. Mucosal Immunol. 2013 doi: 10.1038/mi.2013.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Navarro-Xavier RA, et al. A new strategy for the identification of novel molecules with targeted proresolution of inflammation properties. J. Immunol. 2010;184:1516–1525. doi: 10.4049/jimmunol.0902866. [DOI] [PubMed] [Google Scholar]
  • 33.Chiang N, et al. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature. 2012;484:524–528. doi: 10.1038/nature11042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Spite M, et al. 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]
  • 35. Birnbaum Y, et al. Augmentation of myocardial production of 15-epi-lipoxin-A4 by pioglitazone and atorvastatin in the rat. Circulation. 2006;114:929–935. doi: 10.1161/CIRCULATIONAHA.106.629907. First description of 15-epi-LXA4 biosynthesis by statins in vivo in rats using a novel pathway and phosphorylated enzymes in the biosynthesis of 15-epi-LXA4 that mimic the biosynthetic capacity of acetylated COX-2.
  • 36. Cooray SN, et al. Ligand-specific conformational change of the G-protein-coupled receptor ALX/FPR2 determines proresolving functional responses. Proc Natl Acad Sci U S A. 2013;110:18232–18237. doi: 10.1073/pnas.1308253110. Exciting results indicating that heterodimers and homodimers of ALX/FPR2 signal different responses via activation of distinct intracellular signaling mechanisms. This report is the first demonstration of the enormous flexibility and potential in the receptor ALX for annexin peptide and lipoxin A4, which stimulate IL-10 production and pro-resolving signals.
  • 37.Serhan CN, et al. Novel proresolving aspirin-triggered DHA pathway. Chem. Biol. 2011;18:976–987. doi: 10.1016/j.chembiol.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Morris T, et al. Dichotomy in duration and severity of acute inflammatory responses in humans arising from differentially expressed proresolution pathways. Proc. Natl. Acad. Sci. USA. 2010;107:8842–8847. doi: 10.1073/pnas.1000373107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Brancaleone V, et al. A vasculo-protective circuit centered on lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 operative in murine microcirculation. Blood. 2013;122:608–617. doi: 10.1182/blood-2013-04-496661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Norling LV, et al. Cutting edge: Humanized nano-proresolving medicines mimic inflammation-resolution and enhance wound healing. J. Immunol. 2011;186:5543–5547. doi: 10.4049/jimmunol.1003865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Stables MJ, et al. Transcriptomic analyses of murine resolution-phase macrophages. Blood. 2011;118:e192–e208. doi: 10.1182/blood-2011-04-345330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Miki Y, et al. Lymphoid tissue phospholipase A2 group IID resolves contact hypersensitivity by driving antiinflammatory lipid mediators. J Exp Med. 2013;210:1217–1234. doi: 10.1084/jem.20121887. Identification of a secretory phospholipase A2 activated during contact hypersensitive reactions that is specifically involved in the generation of resolvin D1 and protectin D1.
  • 43.Levy BD, et al. Protectin D1 is generated in asthma and dampens airway inflammation and hyper-responsiveness. J. Immunol. 2007;178:496–502. doi: 10.4049/jimmunol.178.1.496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Miyata J, et al. Dysregulated synthesis of protectin D1 in eosinophils from patients with severe asthma. J Allergy Clin Immunol. 2013;131:353–360. doi: 10.1016/j.jaci.2012.07.048. [DOI] [PubMed] [Google Scholar]
  • 45.Yamada T, et al. Eosinophils promote resolution of acute periotonitis by producing proresolving mediators in mice. FASEB J. 2011;25:561–568. doi: 10.1096/fj.10-170027. [DOI] [PubMed] [Google Scholar]
  • 46.Isobe Y, et al. Stereochemical assignment and anti-inflammatory properties of the omega-3 lipid mediator resolvin E3. J Biochem. 2013;153:355–360. doi: 10.1093/jb/mvs151. [DOI] [PubMed] [Google Scholar]
  • 47.Arita M, et al. Stereochemical assignment, anti-inflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J. Exp. Med. 2005;201:713–722. doi: 10.1084/jem.20042031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ohira T, et al. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J. Biol. Chem. 2010;285:3451–3461. doi: 10.1074/jbc.M109.044131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.El Kebir D, Gjorstrup P, Filep JG. Resolvin E1 promotes phagocytosis-induced neutrophil apoptosis and accelerates resolution of pulmonary inflammation. Proc Natl Acad Sci U S A. 2012;109:14983–14988. doi: 10.1073/pnas.1206641109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Krishnamoorthy S, Recchiuti A, Chiang N, Fredman G, Serhan CN. Resolvin D1 receptor stereoselectivity and regulation of inflammation and pro-resolving microRNAs. Am. J. Pathol. 2012;180:2018–2027. doi: 10.1016/j.ajpath.2012.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Norling LV, Dalli J, Flower RJ, Serhan CN, Perretti M. Resolvin D1 limits polymorphonuclear leukocytes recruitment to inflammatory loci: receptor dependent actions. Arterioscler. Thromb. Vasc. Biol. 2012;32:1970–1978. doi: 10.1161/ATVBAHA.112.249508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fredman G, Li Y, Dalli J, Chiang N, Serhan CN. Self-limited versus delayed resolution of acute inflammation: temporal regulation of pro-resolving mediators and microRNA. Sci Rep. 2012;2:639. doi: 10.1038/srep00639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Li Y, et al. Plasticity of leukocytic exudates in resolving acute inflammation is regulated by microRNA and proresolving mediators. Immunity. 2013;39:885–898. doi: 10.1016/j.immuni.2013.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li D, et al. Resolvin D1 and aspirin-triggered resolvin D1 regulate histamine-stimulated conjunctival goblet cell secretion. Mucosal Immunol. 2013 doi: 10.1038/mi.2013.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Nelson J, et al. The ALX/FPR2 receptor for RvD1 is Expressed and Functional in Salivary Glands. Am J Physiol Cell Physiol. 2013 doi: 10.1152/ajpcell.00284.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jones CN, et al. Microfluidic chambers for monitoring leukocyte trafficking and humanized nano-proresolving medicines interactions. Proc Natl Acad Sci U S A. 2012;109:20560–20565. doi: 10.1073/pnas.1210269109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kasuga K, et al. 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]
  • 58.Simiele F, et al. Transcriptional regulation of the human FPR2/ALX gene: evidence of a heritable genetic variant that impairs promoter activity. FASEB J. 2012;26:1323–1333. doi: 10.1096/fj.11-198069. [DOI] [PubMed] [Google Scholar]
  • 59.Serhan CN, et al. Reduced inflammation and tissue damage in transgenic rabbits overexpressing 15-lipoxygenase and endogenous anti-inflammatory lipid mediators. J. Immunol. 2003;171:6856–6865. doi: 10.4049/jimmunol.171.12.6856. [DOI] [PubMed] [Google Scholar]
  • 60.Hasturk H, et al. 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]
  • 61.Oh SF, Pillai PS, Recchiuti A, Yang R, Serhan CN. Pro-resolving actions and stereoselective biosynthesis of 18S E-series resolvins in human leukocytes and murine inflammation. J. Clin. Invest. 2011;121:569–581. doi: 10.1172/JCI42545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Walker J, et al. Lipoxin A4 increases survival by decreasing systemic inflammation and bacterial load in sepsis. Shock. 2011;36:410–416. doi: 10.1097/SHK.0b013e31822798c1. [DOI] [PubMed] [Google Scholar]
  • 63.Prescott D, McKay DM. Aspirin-triggered lipoxin enhances macrophage phagocytosis of bacteria while inhibiting inflammatory cytokine production. Am. J. Physiol. Gastrointest. Liver Physiol. 2011;301:G487–G497. doi: 10.1152/ajpgi.00042.2011. [DOI] [PubMed] [Google Scholar]
  • 64.Divangahi M, et al. Mycobacterium tuberculosis evades macrophage defenses by inhibiting plasma membrane repair. Nat Immunol. 2009;10:899–906. doi: 10.1038/ni.1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Tobin DM, et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell. 2012;148:434–446. doi: 10.1016/j.cell.2011.12.023. Systematic analysis of TB in infections in zebrafish identify the host gradation response to producing excess leukotriene B4 or excess lipoxin A4, each having different outcomes for the host and the clearance of TB infections.
  • 66.Rajasagi NK, Reddy PB, Mulik S, Gjorstrup P, Rouse BT. Neuroprotectin D1 reduces the severity of herpes simplex virus-induced corneal immunopathology. Invest. Ophthalmol. Vis. Sci. 2013;54:6269–6279. doi: 10.1167/iovs.13-12152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Rajasagi NK, et al. Controlling herpes simplex virus-induced ocular inflammatory lesions with the lipid-derived mediator resolvin E1. J. Immunol. 2011;186:1735–1746. doi: 10.4049/jimmunol.1003456. Exciting first report demonstrating that resolvin E1 protects the eye from Herpes simplex virus infection and stimulates clearance of the virus.
  • 68.Cilloniz C, et al. Lethal dissemination of H5N1 influenza virus is associated with dysregulation of inflammation and lipoxin signaling in a mouse model of infection. J. Virol. 2010;84:7613–7624. doi: 10.1128/JVI.00553-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Morita M, et al. The Lipid Mediator Protectin D1 Inhibits Influenza Virus Replication and Improves Severe Influenza. Cell. 2013;153:112–125. doi: 10.1016/j.cell.2013.02.027. [DOI] [PubMed] [Google Scholar]
  • 70.Tam VC, et al. Lipidomic profiling of influenza infection identifies mediators that induce and resolve inflammation. Cell. 2013;154:213–227. doi: 10.1016/j.cell.2013.05.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Haas-Stapleton EH, et al. Candida albicans modulates host defense by biosynthesizing the pro-resolving mediator Resolvin E1. PLoS ONE. 2007;2:e1316. doi: 10.1371/journal.pone.0001316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Baillie JK, Digard P. Influenza--time to target the host? N Engl J Med. 2013;369:191–193. doi: 10.1056/NEJMcibr1304414. [DOI] [PubMed] [Google Scholar]
  • 73.Pouliot M, Clish CB, Petasis NA, Van Dyke TE, Serhan CN. Lipoxin A4 analogues inhibit leukocyte recruitment to Porphyromonas gingivalis: A role for cyclooxygenase-2 and lipoxins in periodontal disease. Biochemistry (Mosc) 2000;39:4761–4768. doi: 10.1021/bi992551b. [DOI] [PubMed] [Google Scholar]
  • 74.Shen J, et al. Macrophage-mediated 15-lipoxygenase expression protects against atherosclerosis development. J. Clin. Invest. 1996;98:2201–2208. doi: 10.1172/JCI119029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Merched A, 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]
  • 76.Merched AJ, Serhan CN, Chan L. Nutrigenetic disruption of inflammation-resolution homeostasis and atherogenesis. J. Nutrigenet. Nutrigenomics. 2011;4:12–24. doi: 10.1159/000326890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hasturk H, et al. 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]
  • 78. Lima-Garcia J, et al. The precursor of resolvin D series and aspirin-triggered resolvin D1 display anti-hyperalgesic properties in adjuvant-induced arthritis in rats. Br. J. Pharmacol. 2011;164:278–293. doi: 10.1111/j.1476-5381.2011.01345.x. The authors demonstrate that 17-HDHA and aspirin-triggered resolvin D1 have potent protective actions in adjuvant-induced arthritis and reduce pain in this model of arthritis.
  • 79.Kowal-Bielecka O, Kowal K, Distler O, Gay S. Mechanisms of disease: leukotrienes and lipoxins in scleroderma lung disease--insights and potential therapeutic implications. Nat. Clin. Pract. Rheumatol. 2007;3:43–51. doi: 10.1038/ncprheum0375. [DOI] [PubMed] [Google Scholar]
  • 80.Martins V, et al. ATLa, an aspirin-triggered lipoxin A4 synthetic analog, prevents the inflammatory and fibrotic effects of bleomycin-induced pulmonary fibrosis. J. Immunol. 2009;182:5374–5381. doi: 10.4049/jimmunol.0802259. [DOI] [PubMed] [Google Scholar]
  • 81. Borgeson E, et al. Lipoxin A(4) and benzo-lipoxin A(4) attenuate experimental renal fibrosis. FASEB J. 2011;25:2967–2979. doi: 10.1096/fj.11-185017. Expediting resolution and counterregulation of pro-inflammatory mediators by administration of lipoxins A4 or its analogs can head off organ fibrosis and prevent renal fibrosis. An important contribution with far-reaching implications for clinical development of immunoresolvents.
  • 82.Qu X, et al. Resolvins E1 and D1 inhibit interstitial fibrosis in the obstructed kidney via inhibition of local fibroblast proliferation. J Pathol. 2012;228:506–519. doi: 10.1002/path.4050. [DOI] [PubMed] [Google Scholar]
  • 83.Hsiao HM, et al. A novel anti-inflammatory and pro-resolving role for resolvin D1 in acute cigarette smoke-induced lung inflammation. PLoS One. 2013;8:e58258. doi: 10.1371/journal.pone.0058258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang SB, et al. Estrogen negatively regulates epithelial wound healing and protective lipid mediator circuits in the cornea. FASEB J. 2012;26:1506–1516. doi: 10.1096/fj.11-198036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Menon R. The effect of resolvins on dermal wound healing. M.S. thesis. Rutgers; 2012. [Google Scholar]
  • 86. Tang Y, et al. Proresolution Therapy for the Treatment of Delayed Healing of Diabetic Wounds. Diabetes. 2013;62:618–627. doi: 10.2337/db12-0684. Here, the authors demonstrate that pro-resolving mediators such as resolvin D1 can expedite healing of wounds encountered in diabetes.
  • 87.Ramon S, Gao F, Serhan CN, Phipps RP. Specialized proresolving mediators enhance human B cell differentiation to antibody-secreting cells. J Immunol. 2012;189:1036–1042. doi: 10.4049/jimmunol.1103483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hong S, et al. Resolvin D1, protectin D1, and related docosahexaenoic acid-derived products: analysis via electrospray/low energy tandem mass spectrometry based on spectra and fragmentation mechanisms. J. Am. Soc. Mass Spectrom. 2007;18:128–144. doi: 10.1016/j.jasms.2006.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ariel A, 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;280:43079–43086. doi: 10.1074/jbc.M509796200. [DOI] [PubMed] [Google Scholar]
  • 90.Ariel A, et al. Apoptotic neutrophils and T cells sequester chemokines during immune response resolution via modulation of CCR5 expression. Nat. Immunol. 2006;7:1209–1216. doi: 10.1038/ni1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Settimio R, Clara DF, Franca F, Francesca S, Michele D. Resolvin D1 reduces the immunoinflammatory response of the rat eye following uveitis. Mediators Inflamm. 2012;2012:318621. doi: 10.1155/2012/318621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. 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. The authors demonstrate the endogenous biosynthesis of resolvin E1 and resolvin D1 in the retina and the role of cell-cell interactions in this organ.
  • 93.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]
  • 94.Kim TH, Kim GD, Jin YH, Park YS, Park CS. Omega-3 fatty acid-derived mediator, Resolvin E1, ameliorates 2,4-dinitrofluorobenzene-induced atopic dermatitis in NC/Nga mice. Int Immunopharmacol. 2012;14:384–391. doi: 10.1016/j.intimp.2012.08.005. [DOI] [PubMed] [Google Scholar]
  • 95.Hong S, Gronert K, Devchand P, Moussignac R-L, Serhan CN. Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood and glial cells: autacoids in anti-inflammation. J. Biol. Chem. 2003;278:14677–14687. doi: 10.1074/jbc.M300218200. [DOI] [PubMed] [Google Scholar]
  • 96.Lukiw WJ, et al. A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J. Clin. Invest. 2005;115:2774–2783. doi: 10.1172/JCI25420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Marcheselli VL, et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J. Biol. Chem. 2003;278:43807–43817. doi: 10.1074/jbc.M305841200. [DOI] [PubMed] [Google Scholar]
  • 98.Bazan NG, Calandria JM, Serhan CN. Rescue and repair during photoreceptor cell renewal mediated by docosahexaenoic acid-derived neuroprotectin D1. J. Lipid Res. 2010;51:2018–2031. doi: 10.1194/jlr.R001131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Sheets KG, et al. Microglial ramification and redistribution concomitant with the attenuation of choroidal neovascularization by neuroprotectin D1. Mol. Vis. 2013;19:1747–1759. The first description of neuroprotectin D1 action in regulating neovascularization by targeting microglia.
  • 100.Bazan NG, et al. Novel aspirin-triggered neuroprotectin D1 attenuates cerebral ischemic injury after experimental stroke. Exp. Neurol. 2012;236:122–130. doi: 10.1016/j.expneurol.2012.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wang X, et al. Resolution of inflammation is altered in Alzheimer's disease. Alzheimers Dement. 2014 doi: 10.1016/j.jalz.2013.12.024. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mizwicki MT, et al. 1alpha,25-Dihydroxyvitamin D3 and Resolvin D1 Retune the Balance between Amyloid-beta Phagocytosis and Inflammation in Alzheimer's Disease Patients. J Alzheimers Dis. 2013;34:155–170. doi: 10.3233/JAD-121735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Svensson CI, Zattoni M, Serhan CN. Lipoxins and aspirin-triggered lipoxin stop inflammatory pain processing. J. Exp. Med. 2007;204:245–252. doi: 10.1084/jem.20061826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Piomelli D, Sasso O. Peripheral gating of pain signals by endogenous lipid mediators. Nat. Neurosci. 2014;17:164–174. doi: 10.1038/nn.3612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Xu Z-Z, et al. Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions. Nat. Med. 2010;16:592–597. doi: 10.1038/nm.2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bang S, Yoo S, Yang TJ, Cho H, Hwang SW. 17(R)-resolvin D1 specifically inhibits transient receptor potential ion channel vanilloid 3 leading to peripheral antinociception. Br. J. Pharmacol. 2012;165:683–692. doi: 10.1111/j.1476-5381.2011.01568.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Park CK, et al. Resolving TRPV1 and TNF-α-mediated spinal cord synaptic plasticity and inflammatory pain with neuroprotectin D1. J. Neurosci. 2011;31:15072–15085. doi: 10.1523/JNEUROSCI.2443-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Abdelmoaty S, et al. Spinal Actions of Lipoxin A4 and 17(R)-Resolvin D1 Attenuate Inflammation-Induced Mechanical Hypersensitivity and Spinal TNF Release. PLoS One. 2013;8:e75543. doi: 10.1371/journal.pone.0075543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Terrando N, et al. Aspirin-triggered resolvin D1 prevents surgery-induced cognitive decline. FASEB J. 2013;27:3564–3571. doi: 10.1096/fj.13-230276. An exciting discovery demonstrating that resolvin D1 counterregulates pro-inflammatory mediators produced during surgery-induced cognitive decline. Results are the first to demonstrate that resolvin D1 has a protective action in preventing surgery-induced cognitive decline, results that implicate a new approach for perioperative protection and potential for shortening postoperative recovery time.
  • 110.Psychogios N, et al. The human serum metabolome. PLoS One. 2011;6:e16957. doi: 10.1371/journal.pone.0016957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mas E, Croft KD, Zahra P, Barden A, Mori TA. Resolvins D1, D2, and other mediators of self-limited resolution of inflammation in human blood following n-3 fatty acid supplementation. Clin Chem. 2012;58:1476–1484. doi: 10.1373/clinchem.2012.190199. [DOI] [PubMed] [Google Scholar]
  • 112.Serhan CN, et al. 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]
  • 113. Jones ML, et al. Maternal dietary omega-3 fatty acid intake increases resolvin and protectin levels in the rat placenta. J. Lipid Res. 2013;54:2247–2254. doi: 10.1194/jlr.M039842. Important contribution demonstrating resolvins and protectins in placenta and that their levels can be substantially increased with dietary supplementation.
  • 114.Weiss GA, et al. High levels of anti-inflammatory and pro-resolving lipid mediators lipoxins and resolvins and declining docosahexaenoic acid levels in human milk during the first month of lactation. Lipids Health Dis. 2013;12:89. doi: 10.1186/1476-511X-12-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Markworth JF, et al. Human inflammatory and resolving lipid mediator responses to resistance exercise and ibuprofen treatment. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2013;305:R1281–R1296. doi: 10.1152/ajpregu.00128.2013. Exciting new results indicating that strenuous exercise activates acute inflammation and pro-inflammatory eicosanoids that then transitions in humans to pro-resolving mediators present in peripheral blood following a time course consistent with lipid mediator class switching and resolution of exercise-induced muscle stress/inflammation. Importantly, the nonsteroidal anti-inflammatory drug ibuprofen reduces prostaglandin production on strenuous exercise, also reducing leukotrienes and delaying the appearance of pro-resolving mediators, suggesting that nonsteroidal anti-inflammatory drugs delay physiologic resolution following strenuous exercise and thus may have a deleterious effect in muscle physiology.
  • 116. Wu SH, Chen XQ, Liu B, Wu HJ, Dong L. Efficacy and Safety of 15(R/S)-Methyl-Lipoxin A(4) in Topical Treatment of Infantile Eczema. Br J Dermatol. 2013;168:172–178. doi: 10.1111/j.1365-2133.2012.11177.x. A pediatric clinical trial, first to demonstrate that topical addition of aspirin-triggered lipoxin A4 stable analog is safe and effective in reducing infantile eczema.
  • 117.Raatz SK, et al. Baking reduces prostaglandin, resolvin, and hydroxy-fatty acid content of farm-raised Atlantic salmon (Salmo salar) J. Agric. Food Chem. 2011;59:11278–11286. doi: 10.1021/jf202576k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Claria J, Nguyen BT, Madenci A, Ozaki CK, Serhan CN. Diversity of lipid mediators in human adipose tissue depots. Am J Physiol Cell Physiol. 2013;304:C1141–C1149. doi: 10.1152/ajpcell.00351.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Pruss H, et al. Proresolution lipid mediators in multiple sclerosis - differential, disease severity-dependent synthesis - a clinical pilot trial. PLoS One. 2013;8:e55859. doi: 10.1371/journal.pone.0055859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Giera M, et al. Lipid and lipid mediator profiling of human synovial fluid in rheumatoid arthritis patients by means of LC-MS/MS. Biochim Biophys Acta. 2012;1821:1415–1424. doi: 10.1016/j.bbalip.2012.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES