Abstract
Polymorphonuclear leukocyte (PMN)-mediated acute lung injury from ischemia/reperfusion (I/R) remains a major cause of morbidity and mortality in critical care medicine. Here, we report that inhaled low-dose carbon monoxide (CO) and intravenous resolvin D1 (RvD1) in mice each reduced PMN-mediated acute lung injury from I/R. Inhaled CO (125–250 ppm) and RvD1 (250–500 ng) each reduced PMN lung infiltration and gave additive lung protection. In mouse whole blood, CO and RvD1 attenuated PMN-platelet aggregates, reducing leukotrienes (LTs) and thromboxane B2 (TxB2) in I/R lungs. With human whole blood, CO (125–250 ppm) decreased PMN-platelet aggregates, expression of adhesion molecules, and cysteinyl LTs, as well as TxB2. RvD1 (1–100 nM) also dose dependently reduced platelet activating factor-stimulated PMN-platelet aggregates in human whole blood. In nonhuman primate (baboon) lung infection with Streptococcus pneumoniae, inhaled CO reduced urinary cysteinyl LTs. These results demonstrate lung protection by low-dose inhaled CO as well as RvD1 that each reduced PMN-mediated acute tissue injury, PMN-platelet interactions, and production of both cysteinyl LTs and TxB2. Together they suggest a potential therapeutic role of low-dose inhaled CO in organ protection, as demonstrated using mouse I/R-initiated lung injury, baboon infections, and human whole blood.
Keywords: ischemia/reperfusion, resolvins, lung, transcellular eicosanoid biosynthesis, leukotrienes, thromboxane
carbon monoxide (CO) that is produced locally via the hemoxygenase system has emerged as an endogenous gasotransmitter that possesses physiological roles in cardiovascular, immune, and nervous systems (22). Inhaled low-dose CO evokes anti-inflammatory responses both in vivo and in vitro and has a protective function in inflammatory diseases (24). With a systems approach, we recently reported that low-dose inhaled CO accelerates resolution of acute inflammation in mice, enhances human and mouse macrophage phagocytosis and efferocytosis, and temporally regulates local lipid mediators (LM) (7). CO reduces proinflammatory LM and increases specialized proresolving LM (SPM) in mice in vivo and with isolated human macrophages (7). Acute inflammation is fundamentally a protective response to harmful stimuli, such as infection, tissue damage, or irritants. However, when uncontrolled, inflammation can lead to chronic disorders. Resolution of the acute inflammatory response is an active process locally regulated by SPM, which limit further polymorphonuclear leukocytes (PMN) recruitment to the site and enhance uptake of apoptotic PMN and tissue debris by nonphlogistic macrophages and microbial clearance (as reviewed in Refs. 5 and 28). Specific SPM [e.g., resolvin D1 (RvD1), 7S, 8R, 17S-trihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid] activate the endogenous heme oxygenase-1 with local CO production, establishing a proresolving feed-forward circuit (7).
Remote organ ischemia/reperfusion (I/R) is a major challenge during cardiothoracic, vascular, and general surgery (12). I/R is defined as local and remote tissue damage occurring after a transient loss of blood flow and subsequent return (12). During ischemia, inflammatory responses are activated with PMN and platelets that occur first locally. Next, when reperfusion or reflow begins, inflammatory mediators are released. Virtually all organs are subject to remote injury involving leukocyte-mediated tissue damage (11, 12, 23). It is now appreciated that interactions between PMN and platelets lead to transcellular eicosanoid biosynthesis and formation of new mediators that are not produced by either cell type alone (14) that may contribute to events associated with reperfusion.
In the present report, we demonstrated tissue protection with low-dose inhaled CO and Rv that reduce PMN-mediated I/R lung injury. Both CO and RvD1 attenuated PMN-platelet aggregates in murine and human whole blood and reduced leukotrienes (LTs) and thromboxane (Tx) B2. In baboon with Streptococcus pneumoniae (S. pneumoniae)-initiated lung infections, low-dose CO also reduced in vivo cysteinyl LTs, products of PMN-platelet interactions. These results have implications for inhaled low-dose CO as a potential gas treatment in both sterile (I/R) and infection-associated lung injury, that involve disease mechanisms enhanced by PMN-platelet interactions and proinflammatory and bronchoconstrictor eicosanoids such as in sepsis, where new therapeutic approaches are needed.
MATERIALS AND METHODS
I/R induced second-organ reflow injury.
Male FVB mice (6–8 wk; Charles River; LabDiet with essential fatty acids from supplier) were used in accordance with the Harvard Medical Area Standing Committee on Animals (no. 02570 to C. Serhan and no. 04435 to A. Choi). Bilateral hind-limb ischemia was initiated using tourniquets consisting of a rubber band placed on each hind limb. Mice were subjected to hind-limb ischemia for 60 min, after which the tourniquets were removed to initiate reperfusion. CO inhalation was carried out by Laura Fredenburgh, M.D. and Emeka Ifedigbo under an NIH program project (P01-HL108801, A. Choi). Mice were kept in the CO chamber (125–250 ppm) for 60 min before the induction of hind-limb ischemia. RvD1 (250–500 ng with 100 μl of 1% ethanol containing physiological saline; Cayman Chemical) was intravenously administered to the tail vein 5 min before the start of the reperfusion period. Following the reperfusion period (120 min), the mice were euthanized, and their lungs were harvested and stored at −80 C° or in 10% (vol:vol) buffered formalin to be processed for histology assessment by the Histology Core of Boston Children's Hospital. PMN infiltrations into lungs were quantified by lung myeloperoxidase (MPO) as before (29). Briefly, the frozen lungs were homogenized and centrifuged, and the tissue levels of MPO and cytokines were determined with ELISA (R&D Systems and multiplex ELISA). To investigate PMN-platelet aggregates in murine whole blood after I/R, heparinized blood was collected 120 min postreperfusion and incubated with rat anti-mouse CD41 (BD Biosciences, clone MWReg30) and rat anti-mouse Ly-6G (BD Biosciences, clone 1A8) for 30 min at 4°C, followed by red blood cell lysis with 1× RBC lysis buffer (BioLegend) and fixation with 1% (vol:vol) formalin-PBS before flow cytometry with a BD Cant II (BD Biosciences). Data were analyzed using FlowJo (Tree Star).
LM metabololipidomics.
LM metabololipidomics was performed as before (8). Deuterium (d)-labeled internal standards d8-5S-hydoxy-eicosatetraenoic acid (HETE), d4-LTB4, d5-lipoxin (LX) A4, d4-PGE2, d5-RvD2, and d5-LTC4 (500 pg each) in ice-cold methanol were added to facilitate quantification of sample recovery. All samples for LM metabololipidomics were extracted using C18 solid-phase extraction (SPE) columns and subjected to liquid chromatography-tandem mass spectrometry (LC-MS-MS). The LC-MS-MS system, QTrap 6500 (AB Sciex), was equipped with a Shimadzu Prominence LC-20AD HPLC and a Shimadzu SIL-20AC autoinjector (Shimadzu). An Agilent Eclipse Plus C18 column (100 mm × 4.6 mm × 1.8 μm) was used with a gradient of methanol:water:acetic acid of 55:45:0.01 (vol:vol:vol) to 98:2:0.01 at a 0.4 ml/min flow rate. To monitor and quantify the levels of targeted LM, a multiple-reaction monitoring (MRM) method was devised with signature ion fragments for each molecule. Identification was conducted using published criteria (as summarized in Ref. 8) using LC retention times, specific fragmentation patterns, and at least six diagnostic fragmentation ions. Quantification was carried out based on the peak area of the MRM transition, and the linear calibration curves were obtained with an authentic standard for each compound.
Immunofluorescent detection of HIF-1α.
Granulocyte-macrophage colony-stimulating factor-differentiated human macrophages were adhered onto four-well chamber slides overnight (0.1 × 106 cells/well) and incubated with or without 500 ppm CO for 4 h. Cells were then fixed with 1% buffered formaldehyde and permeabilized with 0.1% Triton X-100 for 15 min. After being blocked with 5% FBS containing PBS for 15 min, hypoxia-inducible factor 1α (HIF-1α) was detected by incubation with anti HIF-1α antibody (NOVUS Biologicals, clone H1α67, 1:100 dilution for 1 h at 4°C) followed by incubation with Alexa Fluor 488 goat anti-mouse IgG antibody (Molecular Probes, 1:1,000 dilution for 30 min at 4°C). Nucleus was stained with DAPI (Sigma, Fluoroshield with DAPI). HIF-1α expression and translocation were analyzed using a fluorescent microscope system (Olympus).
Human whole blood incubation.
Human whole blood was drawn from healthy volunteers, who had not taken any medications for 2 wk before donation, by venipuncture in a heparinized syringe (Partners Human Research Committee Protocol No. 1999-P-001297). Human whole blood was incubated with CO (125–250 ppm) at 37°C for 60 min, followed by platelet-activating factor (PAF) stimulation (100 nM, at 37°C for 30 min). Carboxyhemoglobin (CO-Hb) levels were measured by spectrophotometry after reduction with sodium hydrosulfite as before (1). Cell viability was assessed by Trypan blue dye exclusion test after red blood cell lysis with 1× RBC lysis buffer (BioLegend) and by plasma lactate dehydrogenase (LDH) levels (LDH Activity Assay Kit, Sigma) following CO incubation. For evaluation of different agonists, human whole blood was incubated in ambient air or CO (250 ppm) for 60 min (37°C), followed by stimulation at 37°C for 30 min with PAF (100 nM) and U46619 (1 μM) or for 10 min with adenosine diphosphate (ADP) (10 μM) and thrombin (1 U/ml) plus N-Formyl-Met-Leu-Phe (fMLP) (1 μM). To investigate PMN-platelet aggregates and adhesion molecule expressions on PMN and platelet, direct immunofluorescence labeling was performed (at 4°C for 30 min) using anti-human CD41a (Affymetrix eBioscience, clone HIP8), CD18 (BioLegend, clone TS1/18), CD11b (BioLegend, clone CBRM1/5), and CD62P (BD Biosciences, clone AK-4) in combination with the corresponding isotype controls. For LM metabololipidomics, the incubations were stopped with four volumes of ice-cold methanol with deuterium-labeled internal standards and held at −80°C before SPE.
Urinary LTE4 amount in S. pneumoniae-infected baboon and CO treatment.
Baboon sepsis was initiated as before (19). Adult male colony-bred baboons (Papio cynocephalus) were housed in the Duke University Vivarium (Durham, NC) and handled in accordance with American Association for Accreditation of Laboratory Animal Care guidelines. The experimental protocol was approved by the Duke University Institutional Animal Care and Use Committee (A271-11-10). The baboons were sedated, intubated, and mechanically ventilated. The animals were inoculated with S. pneumoniae (Serotype 19A-7; ATCC, 109 colony-forming units per lung), recovered, and extubated. At 48 h under sepsis, urine samples were collected pre-CO exposure. The animals were again sedated, intubated, and treated with inhaled CO (200 ppm) for 60 min under mechanical ventilation. Urine samples were obtained post-CO exposure at 55 h. Urinary LTE4 amount was determined with LC-MS-MS-based metabololipidomics after manual SPE. In brief, columns (SEP-PAK C18, 500 mg, Waters) were equilibrated with 12 ml of methanol and 12 ml of ddH2O. Before extraction, 500 pg of d5-LTC4 was added to 2 ml of urine as an internal standard. After centrifugation at 1,200 g for 10 min, supernatant was acidified (pH = ∼3.5) and immediately loaded onto an SPE column. After being loaded, columns were washed with 12 ml of neutral ddH2O, followed by 10 ml of hexane and 8 ml of methyl formate. Samples were eluted with 8 ml methanol and taken to dryness and resuspended in methanol:water (50:50, vol:vol) for LC-MS-MS-automated injections. Urinary creatinine was measured with creatinine colorimetric assay kit (Cayman Chemical).
Statistical analysis.
Results are expressed as means ± SE. Statistical analysis was performed using the t-test for two-group comparisons and one-way ANOVA test for multiple-group comparisons with post hoc analysis using Dunnett's test (GraphPad Prism). P < 0.05 was considered to be statistically significant.
RESULTS
CO and RvD1 protect lungs from PMN-mediated I/R injury.
We assessed whether inhaled low-dose CO and intravenous RvD1 could each protect from PMN-mediated lung injury following I/R in an established murine model of operating room surgical insults in humans (12, 23). Histology indicated that I/R initiated second-organ lung PMN infiltration (Fig. 1A, top). Low-dose inhaled CO (250 ppm; see Fig. 1) or RvD1 (500 ng/mouse i.v.) each markedly reduced PMN lung infiltration (Fig. 1A, bottom). Combined CO (250 ppm) and RvD1 (500 ng) further decreased lung PMN infiltration compared with each treatment alone. Microscopic examination of the histology demonstrated that mice treated with combined CO and RvD1 showed essentially normal bronchiolar structure. I/R-initiated PMN lung infiltration was quantified by MPO levels (Fig. 1). Inhaled CO dose dependently decreased lung leukocyte influx, giving ∼10% reduction at 125 ppm and ∼55% reduction at 250 ppm (Fig. 1B). For comparison, RvD1 also dampened PMN infiltration, giving ∼70% reduction (Fig. 1C). These results indicated that CO and RvD1 each protected lung from PMN-mediated tissue injury during reflow by reducing PMN infiltration.
Fig. 1.
Carbon monoxide (CO) and resolvin D1 (RvD1) protect lungs from polymorphonuclear leukocyte (PMN)-mediated ischemia/reperfusion (I/R) injury. Mice were subjected to CO inhalation (125–250 ppm; 60 min) before hind-limb ischemia (60 min). RvD1 (250–500 ng) was then administered intravenously followed by reperfusion (120 min). Mice were killed, and lung tissues were collected. A: tissue histology. Hematoxylin and eosin (H&E) staining of I/R lungs (magnification ×40, top, right, ×100). Arrows denote infiltrated PMN. Representative H&E from n = 5 mice from 3 experiments. CO (B) and RvD1 (C) reduction of lung PMN infiltration following I/R. D: reduction in lung PMN influx by CO, RvD1, or both. B–D: PMN infiltration was quantified by lung myeloperoxidase (MPO). Results are MPO values (pg/mg lung tissue; means ± SE, n = 5 mice from 3 experiments). *P < 0.05 vs. I/R alone, †P < 0.05 vs. I/R mice treated with CO (125 ppm) or RvD1 (250 ng) alone.
Next, we tested whether CO and RvD1 could additively protect lung tissues from I/R injury. To this end, mice were treated with 125 ppm inhaled CO, 250 ng RvD1, or a combination of both (Fig. 1D). Subthreshold doses of inhaled CO (125 ppm) or RvD1 (250 ng/mouse i.v.) alone did not significantly reduce PMN tissue infiltration. When administered together, they significantly lowered PMN influx (∼20% reduction), giving additive tissue protection.
CO and RvD1 reduce I/R-initiated LTs and Tx.
LM such as LTB4 and TxB2 play a crucial role in I/R-initiated lung injury (12). Hence, we sought evidence to determine whether CO or RvD1 each regulate LM in I/R injury. Mice were subjected to 60 min of ischemia followed by 120 min of reperfusion, lungs were collected, and LM metabololipidomics performed. Results in Fig. 2A show MRM chromatographs of arachidonic acid (AA)-derived LTs and TxB2 identified in mice lungs. Each LM was profiled using MRM and identified by direct comparison with synthetic and authentic standards using matching criteria, including LC retention time, characteristic fragmentation patterns, and diagnostic ions (Fig. 2B). We quantified each LM identified in lung tissue (Fig. 2C, Table 1) and found that inhaled low-dose CO (250 ppm) and RvD1 (500 ng/mouse) as well as both together gave significant reduction in proinflammatory LTs including LTB4 (∼80% reduction). We also found reduction in TxB2 (∼65% reduction) and cysteinyl LTs with LTC4 and LTE4 (∼85% reductions). These results indicated that CO and RvD1 reduced proinflammatory LTs and Tx in lungs following I/R. Each of these AA-derived bioactive mediators plays crucial roles in pathogenesis of PMN-mediated lung injury (Fig. 2D): LTB4 is a potent PMN chemoattractant, cysteinyl LTs stimulate tissue edema as well as airway smooth muscle contraction, and Tx contributes to platelet activation (25). We monitored a panel of cytokines in lung tissue following hind-limb I/R (Fig. 3) and found that low-dose inhaled CO (250 ppm), RvD1 (500 ng i.v.), and both led to significant reductions of proinflammatory cytokines including interferon (IFN)-γ (∼40%), interleukin (IL)-1β (∼30%), IL-12p40 (∼60%), IL-6 (∼65%), and monocyte chemoattractant protein 1 (∼50%). These results demonstrated that low-dose inhaled CO and RvD1 each reduced proinflammatory cytokine levels in lungs following I/R.
Fig. 2.
CO and RvD1 reduce I/R-initiated leukotrienes (LTs) and thromboxane (Tx). Mice were subjected to I/R second-organ injury (see materials and methods). Lipid mediators (LM) in lungs were identified using LM metabololipidomics. A: representative multiple-reaction monitoring (MRM) chromatographs of the parent ion (Q1) and a diagnostic fragment ion (Q3) in the tandem mass spectrometry (MS-MS). B: representative MS-MS fragmentation patterns and diagnostic ions (insets) employed for identification of LM. C: quantification of lung LM following inhaled CO (250 ppm), administration of RvD1 (500 ng), or both. Results are picograms per milligram of lung tissues; means ± SE, n = 4 mice from 2 experiments. *P < 0.05 vs. I/R alone, †P < 0.05 vs. I/R treated with RvD1 (500 ng). D: arachidonic acid (AA)-derived LTs and Tx pathways. 5-H(p)-ETE, d8-5-hydroxy-eicosatetraenoic acid; 5-LO, 5-lipoxygenase.
Table 1.
Lipid mediators identified in lungs from mice subjected to I/R injury with CO and RvD1 treatments
I/R |
|||||||
---|---|---|---|---|---|---|---|
I/R |
I/R |
CO, 250 ppm |
|||||
Q1 | Q3 | Sham | I/R | CO, 250 ppm | RvD1, 500 ng | RvD1, 500 ng | |
DHA bioactive metabolome and pathway markers | |||||||
RvD1 | 375 | 141 | 0.2 ± 0.1 | 0.6 ± 0.2 | 1.0 ± 0.1 | 1.2 ± 0.1* | 1.7 ± 0.1*† |
PD1 | 359 | 153 | – | – | – | – | – |
Δ15-trans-PD1 | 359 | 153 | 0.3 ± 0.2 | 2.0 ± 0.3 | 0.7 ± 0.2* | 0.5 ± 0.1* | 1.0 ± 0.3 |
10-epi-Δ15-trans-PD1 | 359 | 153 | – | 0.5 ± 0.1 | 0.2 ± 0.1 | 0.3 ± 0.1 | 0.3 ± 0.1 |
10S,17S-diHDHA | 359 | 153 | 1.3 ± 1.2 | 8.1 ± 1.4 | 3.2 ± 1.3 | 2.2 ± 0.5* | 4.6 ± 1.7 |
MaR1 | 359 | 250 | – | – | – | – | – |
Δ12-trans-MaR1 | 359 | 250 | 0.8 ± 0.3 | 1.6 ± 0.8 | 1.3 ± 0.1 | 1.1 ± 0.3 | 0.8 ± 0.1 |
7-epi-Δ12-trans-MaR1 | 359 | 250 | 0.5 ± 0.2 | 0.5 ± 0.1 | 0.2 ± 0.1 | 0.2 ± 0.1 | 0.3 ± 0.1 |
7S,14S-diHDHA | 359 | 250 | 1.1 ± 0.3 | 1.3 ± 0.4 | 0.5 ± 0.1 | 0.5 ± 0.1 | 0.6 ± 0.2 |
EPA pathway markers | |||||||
RvE1 | 349 | 195 | – | – | – | – | – |
RvE2 | 333 | 199 | – | – | – | – | – |
18-HEPE | 317 | 259 | 1.8 ± 0.9 | 15.3 ± 3.2 | 9.5 ± 0.9 | 7.4 ± 0.7* | 12.5 ± 0.7 |
15-HEPE | 317 | 219 | 6.1 ± 4.5 | 57.5 ± 6.5 | 37.4 ± 5.6 | 30.5 ± 2.8* | 50.5 ± 5.4 |
12-HEPE | 317 | 179 | 153.4 ± 91.7 | 605.0 ± 202.3 | 578.0 ± 122.8 | 315.4 ± 117.6 | 591.6 ± 177.6 |
5-HEPE | 317 | 115 | 2.1 ± 1.8 | 8.8 ± 2.9 | 2.6 ± 0.9 | 3.5 ± 0.8 | 4.3 ± 1.3 |
AA bioactive metabolome and pathway markers | |||||||
LXA4 | 351 | 115 | – | – | – | – | – |
LXB4 | 351 | 221 | – | – | – | – | – |
5S,15S-diHETE | 335 | 235 | 5.0 ± 2.0 | 10.3 ± 1.7 | 5.6 ± 1.1 | 4.5 ± 0.3 | 6.0 ± 1.2 |
LTB4 | 335 | 195 | 1.8 ± 0.9 | 9.3 ± 1.3 | 2.7 ± 0.4* | 2.0 ± 0.7* | 2.0 ± 0.3* |
Δ6-trans-LTB4 | 335 | 195 | 4.0 ± 1.3 | 6.0 ± 0.8 | 1.2 ± 0.3* | 1.1 ± 0.2* | 1.0 ± 0.2* |
12epi-Δ6-trans-LTB4 | 335 | 195 | 4.0 ± 1.3 | 5.6 ± 0.6 | 1.2 ± 0.3* | 0.8 ± 0.1* | 1.2 ± 0.3* |
5S,12S-diHETE | 335 | 195 | 3.6 ± 1.6 | 6.4 ± 0.8 | 1.2 ± 0.3* | 1.0 ± 0.3* | 0.9 ± 0.3* |
LTC4 | 626 | 189 | – | 23.2 ± 4.8 | 12.8 ± 2.3* | 5.5 ± 0.9* | 4.4 ± 0.7* |
LTD4 | 497 | 301 | – | – | – | – | – |
LTE4 | 497 | 301 | – | 11.5 ± 1.8 | 4.8 ± 0.5* | 0.8 ± 0.3* | 2.0 ± 1.4* |
PGD2 | 351 | 189 | 50.5 ± 15.6 | 32.0 ± 2.2 | 101.8 ± 23.8* | 19.6 ± 2.4 | 77.1 ± 4.2* |
PGE2 | 351 | 189 | 795.8 ± 638.5 | 2950 ± 450.7 | 3432 ± 186.4 | 2600 ± 352.6 | 3676 ± 208.9 |
PGF2α | 353 | 193 | 181.6 ± 107.6 | 508.4 ± 62.3 | 458.0 ± 46.0 | 351.1 ± 41.9 | 390.7 ± 28.2 |
TxB2 | 369 | 169 | 103.2 ± 39.0 | 326.4 ± 40.3 | 193.2 ± 32.4* | 157.1 ± 13.1* | 118.7 ± 13.8* |
Applicable values are means ± SE of n = 4 mice. Results are expressed as picograms per milligram of lung tissue. Liquid chromatography-tandem mass spectrometry (LC-MS-MS)-based lipid mediator (LM) metabololipidomics were carried out with mice lung homogenates collected at 2 h after reperfusion. The detection limits were approximately 0.1 pg; minus sign denotes below limits of detection. LM were profiled using multiple reaction monitoring (MRM) and identified by direct comparison with synthetic and authentic standards using retention time and at least 6 diagnostic ions. Q1: parent ion, Q3: a diagnostic ion in the MS-MS (daughter ion). Q1-Q3 ion pairs were used for quantification.
P < 0.05 vs. ischemia/reperfusion (I/R) mice.
P < 0.05 vs. resolvin D1 (RvD1)-treated mice.
CO, carbon monoxide; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; AA, arachidonic acid; LX, lipoxin; LT, leukotriene; PG, prostaglandin; Tx, thromboxane.
Fig. 3.
I/R-stimulated cytokines in lungs are reduced by CO and RvD1. Cytokine levels in I/R lungs treated with CO (250 ppm), RvD1 (500 ng), or both. Results are picograms per milligram of lung tissue; means ± SE, n = 5 mice from 3 experiments. *P < 0.05 vs. I/R alone, †P < 0.05 vs. I/R treated with RvD1 (500 ng). MCP, monocyte chemoattractant protein.
Inhaled low-dose CO maintains RvD1 in I/R-injured lungs.
SPM are subject to further local enzymatic metabolism; for example, RvD1 is converted to 8-oxo- and 17-oxo-RvD1 by 15-hydroxy PG dehydrogenase (15-PGDH), an eicosanoid oxidoreductase, which gives dramatically reduced bioactivity (27). CO attenuates 15-PGDH-mediated RvD1 conversion (7). Here, we examined whether inhaled CO has an impact on RvD1 amount in I/R-injured lungs. RvD1 in lungs was profiled using LM metabolomics and identified (Fig. 4 and Table 1). We quantified RvD1 in lung tissue and found that combined CO and RvD1 gave significantly higher RvD1 amounts compared with RvD1 administered alone, suggesting that CO enhances the levels of SPM in I/R lungs by preventing their local inactivation.
Fig. 4.
Inhaled CO maintains RvD1 amount in I/R-injured lungs. RvD1 in I/R-injured lungs was identified using LM metabololipidomics. A: representative MRM chromatograph of the parent ion (Q1) and a diagnostic fragment ion (Q3) in the MS-MS. DHA, docosahexanoic acid. B: representative MS-MS fragmentation pattern and diagnostic ions (insets) employed for identification of RvD1. C: quantification of lung RvD1 amount following inhaled CO (250 ppm), RvD1 (500 ng), or both. Results are picograms per milligrams of lung tissues; means ± SE, n = 4 mice from 2 experiments. *P < 0.05 vs. I/R alone, †P < 0.05 vs. I/R treated with RvD1 (500 ng).
CO and RvD1 reduce PMN-platelet aggregates: mice whole blood after I/R.
I/R injury is known to induce systemic PMN-platelet aggregates in whole blood (12). Hence, we questioned whether CO and RvD1 could each regulate PMN-platelet aggregates during I/R injury. PMN-platelet aggregates in murine whole blood were monitored by expression levels of mouse platelet marker CD41 in the PMN population (Fig. 5, A and B), giving nearly ∼100% increases by I/R compared with sham mice. Low-dose inhaled CO (250 ppm) and RvD1 each significantly reduced PMN-platelet aggregates. Because HIF-1α plays an important role in attenuating acute lung injury (10), we examined the role of CO in regulating HIF-1α in human macrophages. Compared with normoxia, CO 500 ppm (4 h) increased HIF-1α expression and translocation into the nucleus monitored by immunofluorescent staining (Fig. 6). Thus, along with protection of deleterious impact of PMN-platelet aggregation, CO also activated HIF-1α that contributes to lung protection.
Fig. 5.
CO and RvD1 reduce PMN-platelet (Plt) aggregates in mice whole blood after I/R. Mice were subjected to hind-limb ischemia (60 min) and reperfusion (120 min). Whole blood was collected after I/R, and PMN-platelet aggregates were assessed by CD41 expression in PMN population. A: representative density plots of flow cytometry from n = 4 mice from 2 experiments; inset: gate denotes Ly6G+ CD41+ populations. B: results are % Ly-6G+CD41a+ cells representing PMN-platelet aggregates; means ± SE, n = 4 mice from 2 experiments. *P < 0.05 vs. sham control mice, †P < 0.05 vs. I/R alone.
Fig. 6.
CO increases hypoxia-inducible factor-1α (HIF-1α) expression and translocation. Human macrophages were incubated in normoxic condition or with 500 ppm of CO for 4 h. Immunofluorescent detection of cytoplasmic and nuclear HIF-1α in CO-treated group (top) and normoxic group (bottom). Arrowheads indicate protein translocation into nucleus. The scale bar = 50 μm. Results are representative of n = 3 macrophage preparations.
Human translation: CO reduces PMN-platelet aggregates in whole blood.
Next we questioned whether CO could regulate PMN-platelet interactions in human whole blood (15). Fresh human whole blood was incubated in ambient air or CO (125–250 ppm, 60 min, 37°C), followed by addition of PAF (100 nM, 30 min, 37°C). PAF activates both PMN and platelets and has important roles in I/R-stimulated leukocyte adherence (3). To assess potential toxicity of low-dose CO exposure, we monitored CO-Hb, a well-appreciated marker (1), and cell viability with Trypan blue dye exclusion and with plasma LDH after the CO exposure. CO at 250 ppm, the effective inhaled dose, did not alter either CO-Hb or cell viability (Table 2), indicating that, in these incubations, CO was not cytotoxic.
Table 2.
CO-Hb levels and cell viability
Ambient Air | CO Incubation, 250 ppm | |
---|---|---|
CO-Hb, % | 2.4 ± 0.2 | 2.5 ± 0.2 |
Exclusion of Trypan blue staining, % | 96.3 ± 1.2 | 97.3 ± 0.7 |
Plasma LDH, U/l | 74.3 ± 23.2 | 84.7 ± 18.7 |
Results are means ± SE, n = 4 separate healthy donors. Human whole blood was incubated in ambient air or CO (250 ppm) for 60 min (37°C). Carboxyhemoglobin (CO-Hb) levels were measured by spectrophotometric method after reduction with sodium hydrosulfite. Cell viability was assessed by Trypan blue dye exclusion test and plasma lactate dehydrogenase (LDH) levels following CO incubation.
We next monitored expression of adhesion molecules on PMN and platelets, as well as PMN-platelet aggregates, using flow cytometry (Fig. 7, A–C). CO significantly reduced expression of CD11b and CD18 in PMN, with ∼30% and ∼40% reductions at 125 ppm and 250 ppm, compared with ambient air (Fig. 7A). We also assessed platelet CD62P, which were also significantly decreased by CO, giving ∼50% reduction with 125 ppm and ∼60% reduction by 250 ppm, compared with ambient air (Fig. 7B). PMN-platelet aggregates were monitored by expression of CD41a (Fig. 7C) that was reduced ∼40% and ∼50% with CO at 125 ppm and 250 ppm. In separate experiments, RvD1 also dose dependently decreased PAF (100 nM)-induced CD41a in CD18+ PMN populations by 20–25% at 10–100 nM, indicating reduced PMN-platelet interactions (Fig. 7D). In comparison, combination of CO (250 ppm) and RvD1 (10 nM) showed a trend toward further reduction of CD41a compared with each treatment alone but did not prove to be statistically significant (Fig. 7E).
Fig. 7.
In human whole blood, CO reduces PMN-platelet aggregates and adhesion molecules. Human whole blood was incubated in ambient air or CO (125–250 ppm) for 60 min at 37°C, followed by addition of 100 nM platelet-activating factor (PAF) (37°C, 30 min). In select experiments, whole blood was incubated with RvD1 (1–100 nM) for 15 min before addition of PAF. A: PMN CD11b and CD18 expression. B: platelet CD62P expression. C–E: PMN-platelet aggregates monitored by CD41a expression in PMN population. A–C, left: representative histogram plots. Right: % inhibitions by CO. Results are means ± SE, n = 4 (A–D) or n = 3 (E) separate healthy donors. *P < 0.05, **P < 0.01 vs. ambient air incubation without RvD1.
In human whole blood, CO also impacts PMN-platelet aggregates stimulated by specific agonists (Table 3). We selected PAF and thrombin plus fMLP as agonists of both PMN and platelets. The Tx mimetic U46619 and ADP were used as platelet-selective agonists (4, 14). Both PAF (100 nM) and U46619 (1 μM) significantly induced PMN-platelet aggregates (∼220% and ∼110% increases, respectively). ADP (10 μM) and thrombin (1 U/ml) plus fMLP (1 μM) showed ∼80% and ∼75% increases in PMN-platelet aggregates. CO (250 ppm for 60 min) significantly reduced both PAF- and U46619-stimulated PMN-platelet aggregates (∼42% and ∼38% inhibition). In contrast, CO (250–500 ppm for 60 min) did not reduce PMN-platelet aggregates stimulated by either ADP or thrombin plus fMLP (Table 3). We also tested whether CO exposure after stimulation by PAF could reverse PMN-platelet aggregates in human whole blood. To this end, human whole blood was initially stimulated with PAF (100 nM) for 15 min, followed by incubations in ambient air or 250 ppm of CO for 60 min (Table 4). PAF significantly induced PMN-platelet aggregates (∼207% increase) in ambient air incubation. Of interest, in this setting, CO exposure for 60 min after PAF activation of PMN-platelet aggregates in human whole blood showed significant reduction (∼47% inhibition) on these aggregates.
Table 3.
In human whole blood, CO reduces PMN-platelet aggregates: evaluation of agonists
Stimulus | Vehicle (No Stimulus) | Ambient Air + Stimulus | CO + Stimulus | % Reduction by CO |
---|---|---|---|---|
PAF, 100 nM | 546.0 ± 135.4 | 1770.7 ± 253.3* | 1250.9 ± 142.5, 250 ppm† | ∼42% |
U46619, 1 μM | 1166.8 ± 127.8* | 934.8 ± 43.7, 250 ppm† | ∼38% | |
ADP, 10 μM | 630.9 ± 160.8 | 1137.5 ± 64.9* | 1076.3 ± 39.1, 250 ppm | No significant reduction |
1107.9 ± 189.9, 500 ppm | ||||
Thrombin, 1 U/ml | 1105.3 ± 49.8* | 984.8 ± 126.8, 250 ppm | No significant reduction | |
+fMLP, 1 μM | 979.8 ± 73.2, 500 ppm |
Results are means ± SE, n = 4 separate healthy donors. Human whole blood was incubated in ambient air or CO (250 or 500 ppm; 60 min, 37°C), followed by addition of stimuli (37°C, 30 min), including platelet-activating factor (PAF), U46619, adenosine diphosphate (ADP), or thrombin plus N-Formyl-Met-Leu-Phe (fMLP) (10 min). Polymorphonuclear leukocyte (PMN)-platelet aggregates were quantified using flow cytometry (see materials and methods). Results are PMN-platelet aggregates [mean fluorescence intensity (MFI) of CD41a] and % reduction by CO; % reduction was calculated as 100% − {100% × [MFI (CO + stimulus) − MFI (vehicle)]/[MFI (Ambient air + stimulus) − MFI (vehicle)]}.
P < 0.05 vs. vehicle in ambient air incubations.
P < 0.05 vs. respective agonists in ambient air incubations.
Table 4.
In human whole blood, CO reduces preformed aggregates of PMN-platelet
Vehicle | Ambient Air + PAF, 100 nM | CO, 250 ppm + PAF, 100 nM | % Reduction by CO | |
---|---|---|---|---|
PMN-platelet aggregates (MFI of CD41a) | 460.2 ± 149.0 | 1411.4 ± 202.5* | 963.2 ± 272.0† | ∼47% |
Results are means ± SE, n = 3 separate healthy volunteers. Human whole blood was incubated with PAF (100 nM, 37°C) for 15 min allowing PMN-platelet aggregates to form, then incubated in ambient air or CO (250 ppm) for 60 min (37°C). PMN-platelet aggregates were quantified using flow cytometry (see materials and methods). Results are PMN-platelet aggregates (MFI of CD41a) and % reduction by CO; % reduction was calculated as in Table 3, where the stimulus is PAF.
P < 0.05 vs. vehicle in ambient air incubations.
P < 0.05 vs. ambient air incubations.
CO regulates cysteinyl LTs and Tx in human whole blood.
Having found that CO reduced PMN-platelet aggregates, we next questioned whether CO could regulate LM production in human whole blood. Human whole blood was incubated at 37°C in ambient air or CO (250 ppm for 60 min), followed by addition of PAF (100 nM for 30 min). LM in whole blood were profiled using LM metabololipidomics. Figure 8A reports MRM chromatographs of AA-derived TxB2 and cysteinyl LTs identified in human whole blood. Each LM was profiled and identified by direct comparison with synthetic and authentic standards using matching criteria (8) (Fig. 8A). We quantified each LM and found that 250-ppm CO exposure gave 70–80% reduction in PAF-stimulated LTC4, LTD4, and TxB2 (∼90% reduction) in human whole blood (Fig. 8B).
Fig. 8.
In human whole blood, CO regulates cysteinyl LTs and Tx. Human whole blood was incubated in ambient air or CO (250 ppm) for 60 min at 37°C, followed by addition of 100 nM PAF (37°C, 30 min). LM in human whole blood were determined using LM metabololipidomics. A: representative MRM chromatographs of the parent ion (Q1) and a diagnostic fragment ion (Q3) in MS-MS (see materials and methods). Full MS-MS spectra were obtained for each. B: reductions in PAF induced LTC4, LTD4, and TxB2. Results are fold change with CO vs. ambient air; means ± SE, n = 4 healthy volunteers. *P < 0.05 vs. ambient air.
In baboon lung infections, low-dose inhaled CO reduces urinary LTE4 levels.
Systemic inflammation including sepsis produces potent LM, such as cysteinyl LTs, which provoke airway smooth muscle contraction, edema, and further inflammation (21, 25). To assess the extent to which CO regulates cysteinyl LT levels in primates during lung infections, we employed a nonhuman primate (baboon) model of pneumonia (19). Infection was initiated by direct inoculation of S. pneumoniae into both lungs. Diagnosis of baboon sepsis was validated based on hyperthermia, tachycardia, tachypnea, leukocytosis, and positive bacterial culture from peripheral blood (Table 5). These animals met the established criteria for pneumonia (change in white blood cells, signs/symptoms of respiratory infection, and positive bacterial cultures). Urine samples were collected, and the animals were treated with low-dose inhaled CO (200 ppm for 60 min). After inhaled CO, urine samples were collected, and urinary LTE4 was profiled (Fig. 9A) and identified (Fig. 9B). Low-dose inhaled CO gave ∼60% reduction in urinary LTE4.
Table 5.
Physiological and microbiological parameters of Streptococcus pneumoniae-infected baboons
Parameter | Time Point, h | Baboon No. 29223 | Baboon No. 29697 |
---|---|---|---|
Heart rate, bpm | 0 | 94 | 65 |
24 | 132 | 102 | |
48 | 129 | 87 | |
168 | 90 | 78 | |
Temperature, degrees C | 0 | 37.6 | 36.6 |
24 | 39.1 | 38.1 | |
48 | 40.0 | 38.5 | |
168 | 37.9 | 37.1 | |
Respiratory Rate, breaths/min | 0 | 30 | 34 |
24 | 33 | 32 | |
48 | 47 | 36 | |
72 | 24 | 30 | |
96 | 28 | 27 | |
168 | 30 | 30 | |
White blood cell count, ×103 cells/μl | 0 | 19.2 | 7.2 |
24 | 13.0 | 25.3 | |
48 | 23.3 | 16.8 | |
168 | 9.5 | 8.9 | |
% PMNs | 0 | 88.7 | 61.9 |
24 | 86.4 | 96.3 | |
48 | 94.1 | 87.3 | |
168 | 77.9 | 63.5 | |
Blood culture, CFU/ml | 0 | 0 | 0 |
24 | 1 | 0 | |
48 | 0 | 6 | |
168 | 0 | 0 | |
BALF culture, CFU/ml | 0 | 0 | 0 |
48 | 1.20E +05 | 2.00E +03 | |
168 | 0 | 0 | |
Chest radiographs | 0 | Normal | Normal |
48 | Both CXR demonstrate variable bilateral lower lobe opacities |
CFU, colony-forming unit; BALF, bronchoalveolar lavage fluid.
Fig. 9.
Low-dose inhaled CO reduces urinary leukotriene E4 in Streptococcus pneumoniae (S. pneumoniae)-infected baboon. Baboons were sedated, intubated, and mechanically ventilated (17). The animals were bronchoscopically inoculated with S. pneumoniae (109 colony-forming units per lung) to initiate infections via bilateral severe pneumonia. At 48 h, urine was collected pre-CO exposure. The animals were treated with inhaled CO (200 ppm) for 60 min using mechanical ventilation. Urine samples were also obtained after CO treatment (at 55 h). LTE4 in baboon urine were assessed using LM metabololipidomics. A, top: representative MRM chromatographs of the parent ion (Q1) and a diagnostic fragment ion (Q3) in the MS-MS. Bottom: representative MS-MS fragmentation patterns and diagnostic ions (insets) employed for identification of LTE4. B: CO reduced urinary LTE4 in infected baboons. Results are picograms per milligram of urinary creatinine; means ± SE, n = 6 LC-MS-MS chromatographs from n = 2 separate baboons. *P < 0.05 vs. ambient air.
DISCUSSION
In the present report, we demonstrate that low-dose inhaled CO and intravenous RvD1 administration protected against PMN-mediated acute lung injury from I/R (Fig. 1). Lung protection with CO and RvD1 was also commensurate with decreased levels of LTs and Tx in I/R-injured lungs (Fig. 2) and reduction in PMN-platelet aggregates (Fig. 5). The regulation of PMN-platelet interactions in whole blood is of considerable interest in view of the contribution of transcellular production of cysteinyl LTs, which provoke I/R-induced acute lung injury with increased airway contraction and vascular permeability (21, 25). In human whole blood ex vivo, CO exposure inhibited PMN-platelet aggregates and agonist-induced cysteinyl LTs and Tx (Figs. 7 and 8). CO reduction of cysteinyl LTs was also confirmed in vivo during sepsis in baboons (Fig. 9). These results suggest a potential therapeutic value of inhaled low-dose CO in diseases that involve PMN-platelet interactions and excessive production of proinflammatory and bronchoconstrictor (LTC4, LTD4, and Tx) (17, 27) eicosanoids (Fig. 10).
Fig. 10.
Illustration of the mechanisms of action of low-dose inhaled CO and RvD1 on PMN-platelet interactions in lung protection. LOX, lipoxygenase.
I/R-elicited remote tissue injury contributes to morbidity and mortality in a wide range of pathologies, including organ transplantation and cardiothoracic, vascular, and general surgery. Acute lung injury from remote I/R is a serious concern during major surgery (12). We employed an established murine model of remote organ I/R injury (23). Low-dose inhaled CO and intravenous RvD1 administration each protect lung from PMN-mediated tissue injury initiated by I/R (Fig. 1). I/R injury causes sterile inflammation via activation of both innate and adaptive immune responses (12). Uncontrolled systemic inflammation, a sepsis-like state, can develop in sterile inflammation. An example of sterile inflammation resulting in sepsis-like responses includes I/R injury (26, 30). Severe inflammatory stress leads to immune dysfunctions, which are associated with dysregulations of LM profiles (6, 16). During reperfusion, activated PMN and platelets present in systemic circulation become trapped within the microvasculature mainly in lung, where a wide range of proinflammatory chemical mediators are released from activated PMN, platelets, and PMN-platelet aggregates (12). Among them, LTs and Tx are crucial mediators in severe inflammation and have a key role in acute and chronic inflammatory diseases of the cardiovascular and respiratory systems, regulating PMN chemotaxis, edema formation, airway smooth muscle contraction, and platelet activation (Fig. 2) (21, 25). Our present results demonstrated that low-dose inhaled CO and intravenous RvD1 administration reduced proinflammatory LTs and Tx as well as proinflammatory cytokines in lungs following I/R (Figs. 2 and 3). These findings were corroborated with an in vivo baboon infection model that reflects human pathology (19), where the administration of low-dose inhaled CO reduced urinary LTE4, a well-appreciated marker of systemic LT biosynthesis (Fig. 9). For human translation, the CO regulation of both cysteinyl LTs and Tx was agonist selective, reducing PAF-activated human whole blood biosynthesis of LTC4, LTD4, and TxB2 (Fig. 8).
PMN-platelet interactions contribute to transcellular biosynthesis of cysteinyl LTs (Fig. 8) (2, 14). LTA4 within PMN from AA via 5-lipoxygenase is donated to platelets adherent to the neutrophils. Platelets convert LTA4 to LTC4 via LTC4 synthase (14). In murine I/R, there was a twofold induction of PMN-platelet aggregates that were significantly reduced with low-dose inhaled CO and RvD1 (Fig. 5). In addition, CO and RvD1 also significantly reduced cysteinyl LTs in I/R lungs (Fig. 2).
Adhesion molecules have key roles in pathogenesis of I/R injury because they mediate cell-to-cell interactions (12). For example, in remote hepatic injury after gut I/R, CD62P- and CD11/CD18-deficient mice exhibit a blunted leukocyte infiltration to liver (18). It was reported earlier that LXA4 and RvE1 regulate adhesion molecules. LXA4 analogs in nanomolar to micromolar concentrations downregulate CD11/CD18 expression on resting neutrophils, monocytes, and lymphocytes (13). Also, RvE1 in the 10–100 nM range stimulates l-selectin shedding, while reducing CD18 expression in both neutrophils and monocytes in human whole blood (9). In the present study, our results demonstrate that RvD1 (1–100 nM) also dose dependently reduced PAF-stimulated PMN-platelet aggregates in human whole blood (Fig. 7D). Low-dose CO exposure reduced selective agonist (PAF, U46619)-stimulated expression of adhesion molecules on both PMN and platelets and inhibited PMN-platelet aggregate formation in human whole blood (Fig. 7, Table 3) without giving apparent cytotoxicity (Table 2). PAF stimulated Tx formation in human whole blood (Fig. 8). TXA2 initiates the second wave of platelet aggregation and can further increase its own formation via activating phospholipase A2 to liberate free AA that is converted to TXA2 by cyclooxygenase and TXA2 synthase (20). In this regard, CO reduced both PAF and TXA2 agonist U46619-stimulated PMN-platelet aggregation in human whole blood (Table 3), likely via blocking TXA2 synthase, a platelet cytochrome P450 enzyme (4). CO was also shown to inhibit platelet aggregation triggered by ADP and thrombin (4). However, in these experiments, CO did not significantly reduce either ADP or thrombin plus fMLP activation of PMN-platelet aggregates in human whole blood (Table 3). These findings suggest that the dosing of CO in whole blood with ADP or thrombin plus fMLP requires further investigation and that CO inhibition of PAF-induced PMN-platelet aggregation is more easily titratable in human whole blood.
Importantly, low-dose CO exposure reduced preformed PMN-platelet aggregates (Table 4), suggesting a therapeutic potential of low-dose CO to reduce I/R injury via regulating adhesion molecules and PMN-platelet interactions. Of note, hemoxygenase induction produces local endogenous CO that is regulated by resolvins (7), which is relevant as a positive feed-forward loop in organ protection. In summation, we report herein that low-dose inhaled CO and intravenous administration of RvD1 each protected from I/R-mediated acute lung injury. The combination of both displayed additive organ protection. Mechanistically, CO regulated PMN and platelet adhesion molecule expression, as well as reduced PMN-platelet interactions in mice in vivo and human whole blood in vitro. Moreover, the low-dose inhaled CO reduced proinflammatory and bronchioconstrictor eicosanoids (e.g., LTC4, LTD4, and Tx) in I/R mice, specific agonist (PAF and U46619)-stimulated human whole blood, and reduced LTE4 in septic baboons. Taken together, these findings suggest a therapeutic potential for low-dose inhaled CO in regulating disease mechanisms that involve proinflammatory and bronchoactive eicosanoids, PMN-mediated tissue injury, and PMN-platelet interactions in their pathophysiology.
GRANTS
This work was supported in part by NIH grant nos. P01HL108801 and R01GM38765.
DISCLOSURES
C. Serhan is an inventor on patents [resolvins] assigned to the Brigham and Women's Hospital and licensed to Resolvyx Pharmaceuticals. C. Serhan is a scientific founder of Resolvyx Pharmaceuticals and owns equity in the company. C. Serhan'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. No other conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
Author contributions: M.S., M.K., I.R.R., J.D., and B.D.K. performed experiments; M.S., M.K., I.R.R., N.C., J.D., and C.N.S. analyzed data; M.S., J.D., C.A.P., and A.M.C. interpreted results of experiments; M.S. and J.D. prepared figures; M.S. drafted manuscript; N.C., J.D., and C.N.S. edited and revised manuscript; C.N.S. conception and design of research; C.N.S. approved final version of manuscript.
ACKNOWLEDGMENTS
The authors thank M. H. Small for expert assistance in manuscript preparation and J. Lederer (Brigham and Women's Hospital) for multiplex ELISA.
REFERENCES
- 1.Beutler E, West C. Simplified determination of carboxyhemoglobin. Clin Chem 30: 871–874, 1984. [PubMed] [Google Scholar]
- 2.Brady HR, Serhan CN. Adhesion promotes transcellular leukotriene biosynthesis during neutrophil-glomerular endothelial cell interactions: inhibition by antibodies against CD18 and L-selectin. Biochem Biophys Res Commun 186: 1307–1314, 1992. [DOI] [PubMed] [Google Scholar]
- 3.Brancaleone V, Gobbetti T, Cenac N, le Faouder P, Colom B, Flower RJ, Vergnolle N, Nourshargh S, Perretti M. A vasculo-protective circuit centered on lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 operative in murine microcirculation. Blood 122: 608–617, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brune B, Ullrich V. Inhibition of platelet aggregation by carbon monoxide is mediated by activation of guanylate cyclase. Mol Pharmacol 32: 497–504, 1987. [PubMed] [Google Scholar]
- 5.Buckley CD, Gilroy DW, Serhan CN. Proresolving lipid mediators and mechanisms in the resolution of acute inflammation. Immunity 40: 315–327, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chiang N, Fredman G, Backhed F, Oh SF, Vickery T, Schmidt BA, Serhan CN. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature 484: 524–528, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chiang N, Shinohara M, Dalli J, Mirakaj V, Kibi M, Choi AM, Serhan CN. Inhaled carbon monoxide accelerates resolution of inflammation via unique proresolving mediator-heme oxygenase-1 circuits. J Immunol 190: 6378–6388, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Colas RA, Shinohara M, Dalli J, Chiang N, Serhan CN. Identification and signature profiles for pro-resolving and inflammatory lipid mediators in human tissue. Am J Physiol Cell Physiol 307: C39–C54, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dona M, Fredman G, Schwab JM, Chiang N, Arita M, Goodarzi A, Cheng G, von Andrian UH, Serhan CN. Resolvin E1, an EPA-derived mediator in whole blood, selectively counterregulates leukocytes and platelets. Blood 112: 848–855, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Eckle T, Brodsky K, Bonney M, Packard T, Han J, Borchers CH, Mariani TJ, Kominsky DJ, Mittelbronn M, Eltzschig HK. HIF1A reduces acute lung injury by optimizing carbohydrate metabolism in the alveolar epithelium. PLoS Biol 11: e1001665, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med 364: 656–665, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Eltzschig HK, Eckle T. Ischemia and reperfusion–from mechanism to translation. Nat Med 17: 1391–1401, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Filep JG, Zouki C, Petasis NA, Hachicha M, Serhan CN. Anti-inflammatory actions of lipoxin A4 stable analogs are demonstrable in human whole blood: Modulation of leukocyte adhesion molecules and inhibition of neutrophil-endothelial interactions. Blood 94: 4132–4142, 1999. [PubMed] [Google Scholar]
- 14.Fiore S, Serhan CN. Formation of lipoxins and leukotrienes during receptor-mediated interactions of human platelets and recombinant human granulocyte/macrophage colony-stimulating factor-primed neutrophils. J Exp Med 172: 1451–1457, 1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fredman G, Serhan CN. Specialized proresolving mediator targets for RvE1 and RvD1 in peripheral blood and mechanisms of resolution. Biochem J 437: 185–197, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Fullerton JN, O'Brien AJ, Gilroy DW. Lipid mediators in immune dysfunction after severe inflammation. Trends Immunol 35: 12–21, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hamberg M, Svensson J, Hedqvist P, Strandberg K, Samuelsson B. Involvement of endoperoxides and thromboxanes in anaphylactic reactions. Adv Prostaglandin Thromboxane Res 1: 495–501, 1976. [PubMed] [Google Scholar]
- 18.Horie Y, Wolf R, Anderson DC, Granger DN. Hepatic leukostasis and hypoxic stress in adhesion molecule-deficient mice after gut ischemia/reperfusion. J Clin Invest 99: 781–788, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kraft BD, Piantadosi CA, Benjamin AM, Lucas JE, Zaas AK, Betancourt-Quiroz M, Woods CW, Chang AL, Roggli VL, Marshall CD, Ginsburg GS, Welty-Wolf K. Development of a novel preclinical model of pneumococcal pneumonia in nonhuman primates. Am J Respir Cell Mol Biol 50: 995–1004, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Marcus AJ, Safier LB, Ullman HL, Islam N, Broekman MJ, Falck JR, Fischer S, Santos MT, Valles J, von Schacky C. Interactions between platelets and neutrophils in the eicosanoid pathway. Adv Prostaglandin Thromboxane Leukot Res 19: 263–266, 1989. [PubMed] [Google Scholar]
- 21.Martinez Molina D, Wetterholm A, Kohl A, McCarthy AA, Niegowski D, Ohlson E, Hammarberg T, Eshaghi S, Haeggstrom JZ, Nordlund P. Structural basis for synthesis of inflammatory mediators by human leukotriene C4 synthase. Nature 448: 613–616, 2007. [DOI] [PubMed] [Google Scholar]
- 22.Mustafa AK, Gadalla MM, Snyder SH. Signaling by gasotransmitters. Sci Signal 2: re2, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Qiu FH, Wada K, Stahl GL, Serhan CN. IMP and AMP deaminase in reperfusion injury down-regulates neutrophil recruitment. Proc Natl Acad Sci USA 97: 4267–4272, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ryter SW, Choi AM. Gaseous therapeutics in acute lung injury. Compr Physiol 1: 105–121, 2011. [DOI] [PubMed] [Google Scholar]
- 25.Samuelsson B. Role of basic science in the development of new medicines: examples from the eicosanoid field. J Biol Chem 287: 10070–10080, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shen H, Kreisel D, Goldstein DR. Processes of sterile inflammation. J Immunol 191: 2857–2863, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Sun YP, Oh SF, Uddin J, Yang R, Gotlinger K, Campbell E, Colgan SP, Petasis NA, Serhan CN. Resolvin D1 and its aspirin-triggered 17R epimer. Stereochemical assignments, anti-inflammatory properties, and enzymatic inactivation. J Biol Chem 282: 9323–9334, 2007. [DOI] [PubMed] [Google Scholar]
- 28.Tabas I, Glass CK. Anti-inflammatory therapy in chronic disease: challenges and opportunities. Science 339: 166–172, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.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 101: 819–826, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ward PA. New approaches to the study of sepsis. EMBO Mol Med 4: 1234–1243, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]