You are on page 1of 24

Accepted Manuscript

Oral Omega-3 Fatty Acids Promote Resolution in Chemical Peritonitis

Alexander C. Chacon, BSc, Brett E. Phillips, PhD, Miranda A. Chacon, BSc, Deborah
Brunke-Reese, BSc, Shannon L. Kelleher, PhD, David I. Soybel, MD

PII: S0022-4804(16)30157-3
DOI: 10.1016/j.jss.2016.06.036
Reference: YJSRE 13834

To appear in: Journal of Surgical Research

Received Date: 5 February 2016


Revised Date: 1 June 2016
Accepted Date: 10 June 2016

Please cite this article as: Chacon AC, Phillips BE, Chacon MA, Brunke-Reese D, Kelleher SL, Soybel
DI, Oral Omega-3 Fatty Acids Promote Resolution in Chemical Peritonitis, Journal of Surgical Research
(2016), doi: 10.1016/j.jss.2016.06.036.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
Revised 05-25-2016

Oral Omega-3 Fatty Acids Promote Resolution in Chemical Peritonitis

Short title: Oral Omega-3s in Chemical Peritonitis

PT
Alexander C. Chacon, BScd, Brett E. Phillipsa, PhD, Miranda A. Chacon, BScd,

RI
Deborah Brunke-Reese, BSca , Shannon L. Kellehera,b,c, PhD, and David I. Soybel, MDa,b

SC
Departments of Surgerya, Cellular and Molecular Physiologyb, and Pharmacologyc, Pennsylvania State

University College of Medicined, Hershey, Pennsylvania, USA.

U
AN
*Corresponding Author:
M

David I. Soybel MD
Department of Surgery
D

Penn State College of Medicine


500 University Drive
TE

PO Box 850, Mail Code H149


Hershey, PA 17033
Tel 717-531-5272
FAX 717-531-0884
EP

Email dsoybel@hmc.psu.edu
C

Authors’ contributions: A.C.C., B.E.P., S.L.K., and D.I.S. designed the study. A.C.C., B.E.P., M.A.C., and
AC

D.B-R. executed the experiments. A.C.C., B.E.P., and D.I.S. interpreted the data and wrote the

manuscript. All authors discussed, revised the manuscript for intellectual content, and gave approval of

the final version.

Disclosures: A.C.C., B.E.P., M.A.C., D.B-R., S.L.K., and D.I.S. reported no proprietary or commercial

interest in any product mentioned or concept discussed in the article.


ACCEPTED MANUSCRIPT
2

Abstract

Introduction

Recent studies suggest that purified omega-3 fatty acids may attenuate acute inflammation and hasten

the transition to healing. In this study, we tested the hypothesis that pre-treatment with omega-3 rich

PT
fish oil (FO) would promote resolution of peritoneal inflammation through production of specific lipid

RI
mediators.

Methods

SC
C57/BL6 mice were given a daily 200 μL oral gavage of saline (CTL) or FO (1.0-1.5 g/kg/d DHA and 1.3-2.0

g/kg/d EPA) for 7 days before chemical peritonitis was induced with thioglycollate. Peritoneal lavage

U
fluid was collected before induction and at days 2 and 4 post peritonitis onset. Prostaglandin E2 (PGE2),
AN
Leukotriene B4 (LTB4), Resolvin D1 (RvD1), and the composition of immune cell populations were

examined in peritoneal lavage exudates. Cells harvested from the peritoneum were assessed for
M

macrophage differentiation markers, phagocytosis, and LPS-induced cytokine secretion profiles (IL-6, IL-
D

10, IL-1β, TNFα).


TE

Results

The ratio of RvD1 to pro-inflammatory PGE2 and LTB4 was increased in the peritoneal cavity of FO
EP

supplemented animals. FO induced a decrease in the number of monocytes in the lavage fluid, with no

change in the number of macrophages, neutrophils, or lymphocytes. Macrophage phagocytosis and


C

M1/M2 mRNA markers were unchanged by FO with the exception of decreased PPARγ expression. FO
AC

increased ex vivo TNFα secretion after stimulation with LPS.

Conclusions

Our findings provide evidence that nutraceutically-relevant doses of FO supplements given before and

during chemical peritonitis shift the balance of lipid mediators towards a pro-resolution, anti-
ACCEPTED MANUSCRIPT
3

inflammatory state without drastically altering the number or phenotype of local innate immune cell

populations.

Keywords: Fish Oil, Inflammation, Resolution, Peritonitis, Resolvin D1, Leukotriene B4

PT
RI
U SC
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
4

Introduction

Resolution of local and systemic inflammation is critical in the process of recovery from

peritonitis. Once thought to be a passive process involving depletion of pro-inflammatory mediators,

resolution is now known to be an active, cell-mediated progression involving the actions of specific lipids

PT
and proteins as drivers of the resolving response. Novel discoveries 1-3 have illuminated the second half

RI
of this bimodal response to immune challenge, which is dictated in part by bioactive lipid mediators with

varied functions. As reviewed elsewhere4, pro-resolution lipid mediators - resolvins, maresins and

SC
protectins – appear during the first phase of inflammation and are capable of inducing the resolution

program, which includes the eventual deceleration of recruitment of neutrophils and acceleration of

recruitment of macrophages to inflamed spaces3,5.

U In the resolution phase, the profile of the


AN
macrophage population is switched from M1 (pro-inflammation) to M2 (pro-resolution)6, which includes

decreased production and/or down-regulation of pro-inflammatory cytokines7,8 and alterations in


M

immune cell shape and morphology consistent with the scavenging of non-viable cells and material, a

process known as efferocytosis9. It has been postulated that altered ratios of pro-resolution to pro-
D

inflammation lipids (prostaglandins, leukotrienes) may effectively dictate the character of an


TE

inflammatory response by shifting the balance of pro- and anti-inflammatory cytokines and
EP

chemokines10.

Endogenous production of pro-resolving lipid mediators depends on availability of specific


C

precursors in the omega-3 (n-3) fatty acid class, including DHA (docosahexaenoic acid) and EPA
AC

(eicosapentaenoic acid). Thus, it has been hypothesized that resolution of certain states of chronic

inflammation might be interrupted by treatment with these compounds, either in purified form or in the

form of fish oil (FO) supplements. As the FO industry has grown to become a nearly $2 billion market

and recent surveys have shown that 1/3 of Americans taking nutritional supplements may be using a FO

supplement11, the need exists to carefully examine how omega-3 administration may affect medical
ACCEPTED MANUSCRIPT
5

care, especially in relation to invasive procedures. Indeed, the benefits of purified and dietary

supplement forms have been observed in some settings associated with the surgical patient. Recent

meta-analyses show that enteral immunonutrition regimes, which include omega-3 supplements, may

optimize recovery of patients who undergo elective surgery for GI cancer, especially when delivered in

PT
the perioperative period12,13, while some parenteral models of omega-3 supplementation have also

shown beneficial effects14,15. In murine models a similar mixed picture exists, but in congruence with

RI
human models a majority of studies providing FO supplementation over periods of 5 days to 12 weeks

SC
display downstream resolution-favoring changes in mRNA expression of classical M1/M2 macrophage

markers of maturation16, inflammatory cytokine and lipid profile17-19, efferocytosis20, and immune cell

U
oxidative potency6, with additional reports indicating that oral FO takes 24-72 hours to induce beneficial
AN
metabolic effects7.

It remains unclear what sorts of benefits would be observed in settings of more acute
M

inflammation such as recovery from surgery 21,22. Critical concerns in design of such studies include the
D

demonstration that FO supplementation actually results in increased production of pro-resolution lipid

mediators and that they are available in the inflamed space. Also important is the need to characterize
TE

changes in profiles of pro- and anti-inflammation cytokines resulting from such supplementation23-25.
EP

Lastly, it remains unclear whether dietary supplements can elicit the phenotypic switch to resolution

with efficacy similar to that of purified forms of DHA/EPA but with less toxicity.
C

In this study, we utilized a mouse model of sterile chemical peritonitis to characterize the local
AC

transition from acute inflammation to resolution in response to dietary FO supplements that may be

obtained commercially. Thioglycollate is a relatively mild chemical irritant which provides a

reproducible and consistent approach to study cascades of inflammation and resolution in the mouse

and rat analogous to those in chemical and mechanical peritonitis conditions such as gastric acid leak,

abdominal trauma, and surgical bowel irritation, without resulting in the presence of confounding
ACCEPTED MANUSCRIPT
6

polymicrobial infection26. Peritoneal injection of thioglycollate has been utilized as a low mortality, low

systemic sequelae method to recruit and subsequently harvest inflammation-driving resident

macrophages for the purposes of in vitro and ex vivo study, and captures cells transitioning between

acute inflammation and active resolution (3-4 days post-injection) with peak inflammatory response in

PT
the first 1-2 days and complete clearance by day 7-827. Using this model, we find that relevant doses of

RI
oral FO favor production and local appearance of resolution-phase lipid mediators but may yet be

unable to elicit the dramatic changes in cell number and cytokine production that would be required to

SC
effect a clear benefit in accelerating recovery from an acute sterile inflammatory insult.

Materials/Methods

U
AN
Animals and Experimental Design

All animal procedures were approved and conducted in accordance with the Institutional Animal
M

Care and Use Committee of Penn State Milton S. Hershey College of Medicine. Male C57BL/6 mice were
D

obtained commercially (Taconic, Hudson, NY) and individually housed in polycarbonate cages with a 12

h light/dark cycle. Solely male mice were utilized, as recent reports have displayed differences in
TE

leukotriene biosynthesis between sexes following chemical peritonitis28. Animals were fed ad libitum
EP

with the commercial AIN-93M (MP Biomedicals, Santa Ana, CA) diet containing 4% soybean oil for 28

days starting at 10-12 weeks of age. On days 21 through 28, mice were given 200 µL daily oral gavages
C

of PBS as control (CTL) or Marinol C-38 (FO) (Stepan, Maywood NJ), a proprietary fish oil blend
AC

containing 17% DHA and 24% EPA (approximately 1.0-1.5 g/kg/d DHA and 1.3-2.0 g/kg/d EPA). At day

28, mice from each group were either euthanized to establish baseline infiltrative and secretory

characteristics of immune cell populations within the peritoneum (Day 0) or 1mL 3% thioglycollate was

injected into the peritoneal cavity to induce a chemical peritonitis, as established in prior studies27.

Mice given thioglycollate were euthanized at day 2 or 4 post-injection. Mouse diets and oral gavages
ACCEPTED MANUSCRIPT
7

were continued throughout the additional duration of chemical peritonitis. Once euthanized, mouse

blood was collected via cardiac puncture and plasma was isolated via centrifugation at 1000 rcf and

immediately flash-frozen in liquid nitrogen. Additionally, the peritoneal cavity was rinsed with 5mL cold

1X PBS, the peritoneal lavage exudate was collected, and the cellular and liquid exudate fractions were

PT
isolated via centrifugation at 3000 rcf. The supernatant was flash-frozen.

RI
Ex vivo cytokine release and bioparticle phagocytosis

Peritoneal exudate cells from 6-8 mice per group were isolated and counted via

SC
hemocytometry. 2.5X105 cells from mice 2 and 4 days post-injection were suspended in 200 µL media,

plated on 96 well plates and incubated for 16 h with and without LPS (100 ng/mL). Cells from day 0 mice

U
at baseline were not utilized as cell numbers were too low for all assays except for cytologic analysis.
AN
Following stimulation, culture media was removed and assessed for cytokine content (IL-6, IL-10, IL-1β,

and TNFα) by ELISA according to the manufacturer’s protocols (Duoset, R&D Systems, Minneapolis, MN).
M

Immune cell phagocytosis was assessed on the same cells via incubation with fluorescent E. coli particles
D

(Vybrant Phagocytosis Assay Kit (Life Technologies, Frederick, MD)) for 2 h, after which intracellular

fluorescence was measured in a Synergy-2 fluorescent plate reader at 480 nm excitation and 520 nm
TE

emission settings.
EP

Cytology of infiltrated exudate cells

1X105 exudate cells from mice at baseline or at 2 and 4 days post-injection were independently
C

plated on Superfrost microscope slides using a CytoPro centrifugation system, allowed to dry for 1 h,
AC

and fixed with 50 μL MeOH. Slides were stained with 50 μL Wright/Giemsa stain to differentiate

immune cell types. Briefly, on one slide per mouse and 4 field counts per slide, cell types were

determined according to cell morphology, nuclear characteristics, cytoplasmic contents, and color. Cells

were deemed vacuolar if more than 50% of the cytoplasm consisted of vacuoles. To ensure objective

determination of cell morphology and identification of cellular debris, two individuals independently
ACCEPTED MANUSCRIPT
8

assessed unidentified samples (results averaged) to allow for the novel characterization of macrophage

vacuole content.

Gene expression profiling by quantitative PCR

Cells harvested 4 days post-thioglycollate injection (90-95% macrophages) were assessed for

PT
macrophage differentiation markers via quantitative PCR (qPCR). RNA was isolated with the Totally RNA

RI
kit (Ambion, Naugatuck, CT) and 1 µg was used to generate cDNA with the ImProm-II Reverse

Transcription System (Promega, Madison, WI). qPCR was performed with the QuantStudio 12K Flex

SC
machine (ThermoFisher, Waltham, MA) using the FastStart Universal SYBR Green Master (ROX) mix

(Roche, Mannheim, Germany). All primers for Actin29, Arginase 130, CD3629, IL-630, iNOS30, PPARγ31,

U
Ym230, and 12/15 LO32 were purchased from Integrated DNA Technologies (Coralville, IA), and consisted
AN
of sequences utilized in the literature.

Peritoneal exudate lipid assessment


M

Frozen peritoneal exudate supernatant was assessed for Prostaglandin E2 (PGE2), Leukotriene
D

B4 (LTB4) and Resolvin D1 (RvD1) content. Lipid fractions of the exudate supernatant were

concentrated using the Folch method33. Briefly, 2mL of sample was extracted using 8mL 2:1
TE

chloroform/methanol, vortexed thoroughly in polystyrene tubes, incubated 5 min, and centrifuged at


EP

4500 rpm for 20 min (room temperature). The organic (lower) phase was collected using sterile glass

pipettes and concentrated via vacuum centrifuge. Lipid content of isolated samples was assessed by
C

ELISA according to company specifications (Cayman Chemical, Ann Arbor, MI). Total protein content in
AC

the exudate supernatant was assessed via Bradford Reagent (Sigma-Aldrich, St Louis, MO) and was

unchanged between treatment conditions (data not shown).

Statistical Analysis

Data are presented as means ± SEM. Data was assessed by Student t-test for comparison of

control and fish oil groups, or by Two-way ANOVA to examine treatment and time point interactions
ACCEPTED MANUSCRIPT
9

using GraphPad Prism, version 5.03. Bonferroni post-tests were run to examine significant effects of

time. A p-value of <0.05 was considered significant.

Results

PT
Supplement impact on functional parameters

No observable differences in alertness, grooming habits, posturing, or skin health of mice

RI
administered PBS or FO was noted. Basic assessments of mouse vitality, including baseline weight (the

SC
start of AIN 93 diet), weight change over the experimental period, and food intake were not different

between the groups (Table 1).

U
Effect of Fish Oil on Immune Cell recruitment
AN
Recruitment of immune cells in response to chemical peritonitis in CTL and FO mice was determined

via Wright-Giemsa staining and differential classification. Thioglycollate induced significant recruitment
M

of immune cells at days 2 and 4 (Figure 1). Monocyte number was significantly reduced with FO
D

supplementation compared to control mice, p<0.05. Monocyte content was greater on day 2 than on

days 0 and 4. Cells derived from the monocyte lineage with mature characteristics (macrophages/
TE

[monocytes + macrophages]) were significantly increased with FO supplementation. FO did not


EP

significantly alter the total number of cells found in the peritoneal lavage before or after induction of

peritonitis with 3% thioglycollate. There were no significant differences in the numbers of macrophages,
C

neutrophils, lymphocytes, plasma cells (Figure 1) or basophils, eosinophils, mast cells, or dead/dying
AC

cells in the peritoneal lavage (data not shown). It was observed that macrophages exhibited a more

vacuolar morphology following FO supplementation, though this effect was not significant, p=0.23.

Supplement impact on cytokine production, phagocytosis, and mRNA expression

Ex vivo cytokine secretion profiles in response to the danger signal LPS were examined in peritoneal cells

isolated on days 2 and 4. 16 h of LPS treatment induced a marked increase in secretion of IL-6, TNFα, IL-
ACCEPTED MANUSCRIPT
10

10, and IL-1β (Figure 2) from baseline, which was nearly undetectable without stimulation (data not

shown). Interestingly, TNFα secretion with LPS stimulation was greater in the FO group than the CTL

group, p<0.01. In both treatment conditions, secretion of IL-10 decreased and TNFα secretion increased

over time from day 2 to day 4, while IL-6 and IL-1β levels remained unchanged from day 2 to day 4

PT
regardless of treatment.

RI
To examine classical or alternative macrophage activation in response to chemical immune

challenge with or without FO supplementation, expression profiles of peritoneal cells at day 4 (90-95%

SC
macrophages) were determined via qPCR. PPARγ mRNA levels were significantly decreased with FO

supplementation, p<0.01 (Figure 2e). FO did not induce any differences in mRNA levels of Arginase 1,

U
CD36, IL-6, iNOS, Ym2, or 12/15 LO in the recruited peritoneal immune cells.
AN
Ex vivo E. coli bioparticle phagocytosis was examined in peritoneal cells isolated on days 2 and 4

(Figure 2f). There were no significant differences in phagocytosis by recruited immune cells.
M

Effect of fish oil on lipid mediator production


D

Lipid mediators of inflammation were assessed in the peritoneal lavage. The ratio of anti-

inflammatory (RvD1) to pro-inflammatory (PGE2 or LTB4) lipid mediators was examined, as the balance
TE

between these two groups establishes the immune response. FO supplementation increased the
EP

RvD1/PGE2 ratio, RvD1/LTB4 ratio, and RvD1/ (PGE2+LTB4) ratio (Figure 3), p<0.05. Additionally, FO

reduced concentration of LTB4 in the peritoneal lavage (p<0.05), but elicited no significant change in
C

RvD1 (p=0.16) or PGE2 (p=0.18). The concentration of LTB4 was elevated on day 2 as compared to days
AC

0 and 4. RvD1/LTB4 and RvD1/(PGE2 + LTB4) ratios were elevated on day 0 compared to days 2 and 4.

Discussion

The potential for specific natural and synthetic pro-resolving lipid compounds to alleviate

inflammation and hasten recovery from injury and illness has been well established in recent years34,35.
ACCEPTED MANUSCRIPT
11

Administration of highly processed nutraceutical agents may, however, be attended by unanticipated

toxicities or undesirable pharmacologic effects at high doses, with undetermined safety and utility of FO

supplementation pre and peri-operatively. Fish oil appears to be safe at doses up to 7 g/d, even in

patients on anticoagulant therapy, but may induce bleeding at doses of near 20 g/d in humans36, and the

PT
side effect profile of sub-acute or chronic supraphysiologic omega-3 fatty acids or targeted pro-resolving

RI
medicines is uncertain. Additionally, the translatability of studies incorporating pro-resolving lipids as

targeted agents, including the physiology dictating their effects on healing and convalescence in surgical

SC
illness, remains unclear. In this study of resolution from chemical peritonitis in mice, we demonstrated

that 7 days of oral fish oil gavage at a dose of approximately 6-9 g/kg/d (1.0-1.5 g/kg/d DHA and 1.3-2.0

U
g/kg/d EPA) could increase the ratio of resolution phase lipid mediators to inflammatory lipid mediators
AN
while inducing only subtle alterations in immune cell type and number, cytokine production, and

functional status.
M

As others have shown no change in Resolvin D1 content in serum37 and only modest changes in

D and E series Resolvins in the bone marrow following dietary omega-3s in comparable doses38, we
D

sought to assess changes in inflammatory cell types and function in a specific, localized space to better
TE

understand transitions from inflammation to resolution in peritonitis. In response to oral omega-3


EP

supplementation, our data display a decrease in the number of monocytes present in the mouse

peritoneum following thioglycollate-induced chemical peritonitis, with no change in neutrophil or


C

lymphocyte number. This stands in contrast to the results of prior chemical peritonitis studies in mice
AC

that have used intraperitoneal or intravenous administration of pro-resolution DHA and DHA- or EPA-

derived lipid mediator products, where moderate reductions in neutrophil influx39 as well as alterations

in efferocytotic capacity40-45 were noted. We also observed an increase in the ratio of macrophages/

(monocytes + macrophages) in the peritoneal space in mice supplemented with fish oil, with no change

in the number of macrophages present. This result indicates that cells derived from the monocytic cell
ACCEPTED MANUSCRIPT
12

line are present in the space following omega-3 supplementation; however, infiltrated or locally derived

early phase monocytic cells either do not migrate into the peritoneum in response to chemical challenge

or do not remain undifferentiated in the space for long before transitioning to macrophages. We did

note that cells from mice supplemented with fish oil were more vacuolar appearing, especially on day 2

PT
post injection; however, this result was not statistically significant and no change in bioparticle

RI
phagocytosis was observed in response to fish oil supplementation. Together, these results

demonstrate that oral FO supplementation does result in alterations in inflammatory cell recruitment

SC
and maturation in response to chemical peritonitis, but that this effect does not mimic the more potent

functional and phenotypic changes seen with administration of the downstream resolvins, maresins, and

protectins.

U
AN
In this study, we demonstrate that oral fish oil supplementation induced a significant increase in

TNFα secretion from isolated exudate cells challenged with LPS. This result is produced in a model of
M

secondary macrophage activation, differing from models assessing systemic TNFα burden or models

assessing co-incubation of cells with resolvins and LPS42,46. Moreover, this effect is observed 2 and 4
D

days downstream of in vivo chemical insult, perhaps indicative of retained ability of macrophages to
TE

elicit their cell recruitment and activation functions. Furthermore, in our day 4 exudate cellular
EP

population (90-95% macrophages), PPARγ mRNA expression was 27% lower in mice supplemented with

FO, with a limited response in mRNA message for classical M1/M2 phenotypic markers of macrophage
C

differentiation. In vitro studies assessing the relationships of PPARγ lipid ligands, PPARγ binding, and
AC

downstream upregulation of macrophage differentiation markers such as CD36 (a surface receptor

implicated in efferocytosis) and Arginase 1 (a transaminase implicated in the classical M1/M2 switch)

have begun to uncover some as-of-yet unclear associations17,29,47-49. Akin to our results, one study

displayed a 26% decrease in PPARγ mRNA in mouse aortas following a 12 week fish oil diet; in another

study TNFα decreased PPARγ synthesis in macrophages50, perhaps providing a mechanism for the
ACCEPTED MANUSCRIPT
13

decreased PPARγ mRNA, unchanged particle phagocytosis and unchanged macrophage vacuolization in

this study. These results imply solely a mild change in M1/M2 macrophage phenotype occurs after

administration of short-term oral FO supplementation, in contrast to the potent effects of resolution-

phase lipid mediators6,42,46,51

PT
Our mouse model of FO supplementation displayed an increase in the ratio of pro-resolving

RI
RvD1 to pro-inflammatory LTB4 and PGE2, a novel finding which may indicate a relevant shift in

inflammatory balance favoring milder inflammation and improved resolution. This effect was not

SC
attributable to Resolvin D1 alone as it only displayed a trend of increasing concentration with FO

supplementation. Here, LTB4 decreased in response to FO. These findings suggest that the dietary

U
omega-3 fatty acids DHA and EPA promote a pro-resolution lipid mediator milieu not only through the
AN
distinct resolution-phase actions of their downstream precursors, but also through suppression of

previously active inflammatory lipid cascades. LTB4 production is hindered by increasing pro-resolving
M

metabolite concentrations51,52, and it has been shown that diminished LTB4 production results in

diminished neutrophil influx into the peritoneum in surgically induced bacterial peritonitis in mice53. We
D

propose that therapies which increase the RvD1 to LTB4 ratio may hasten the recovery from
TE

inflammatory surgical illness.


EP

In conclusion, we have shown that relevant doses of FO in the mouse can induce a pro-resolving

lipid mediator milieu in the peritoneum without severely altering the recruitment of immune cells to the
C

peritoneal space. This occurs during the transition phase from acute inflammation to active resolution.
AC

These results contrast those from studies utilizing purified lipid mediators, which have displayed great

potency in mouse and human models of inflammation. Further characterization of ratios between lipid

mediators in various disease states still remain to be identified, and approaches developed to intervene

are experimental. Many studies addressing the effects of resolvins, lipoxins, protectins, and maresins in

resolution from inflammatory conditions are underway, though basic dietary studies using whole fish oil
ACCEPTED MANUSCRIPT
14

and precursor lipid compounds should not be abandoned until more is known about lipid mediator

biology in surgical diseases.

In the setting where millions of Americans are already consuming dietary or supplemental fish

oil of their own accord, it is possible that under certain clinical circumstances oral preparations of lipid

PT
precursors such as DHA and EPA will result in mild to moderate accumulation of pro-resolving lipid

RI
mediators, with still unclear consequences on convalescence from surgery. This study provides a

foundation for understanding lipid and macrophage biology in acute inflammation in a controlled and

SC
non-infectious environment, helping to clarify the results of microbe-dependent pre-clinical models

studying omega-3 administration in bowel perforation, bacterial peritonitis, sepsis, and ileus. Additional

U
studies of chemical peritonitis incorporating concomitant mono- or poly-microbial infection are
AN
warranted in order to fully understand the role that omega-3 supplementation may play in the

management of most clinical peritonitis scenarios. As others have shown, pre-, peri-, or post-
M

operatively there may be opportunities to improve healing and convalescence using omega-3 therapies

without increasing the risk of post-operative surgical site complications and infection14, 54, 55, 56.
D
TE

Acknowledgements:
EP

E. coli bacterial endotoxin was a gift of Dr. James A. Lederer, (Brigham and Women’s Hospital, Boston,

MA). This study was funded by the Pennsylvania State University College of Medicine Department of
C

Surgery.
AC

Figure Legends

Cover Legend. Representative Wright-Giemsa stain of cells isolated from mouse peritoneal lavages 2
days post-induction of chemical peritonitis, visualized via light microscopy. Slide displays monocytes,
macrophages, neutrophils, and lymphocytes from peritoneal lavages. Mice were treated with 7 days of
saline or fish oil oral gavage prior to induction of peritonitis with thioglycollate.
ACCEPTED MANUSCRIPT
15

Figure 1. Influence of FO supplementation on peritoneal immune cell populations recruited during


peritonitis. Wright-Giemsa stains of cells isolated from mice at day 0, 2, and 4 post-thioglycollate
injection were visualized via light microscopy for CTL mice (a, b, and c respectively) and FO mice (d, e, f).
Cells were classified by morphologic features and quantified. White bars indicate CTL, black bars
indicate FO. Results are presented as means ± SEM, with statistically significant treatment effects
denoted by * (2-way ANOVA, p < 0.05). Significant effects of time via 2-way ANOVA are not displayed
due to low cell content on day 0; effects due to time on days 2 and 4 are displayed based on Bonferroni

PT
post-tests signified by letters. The number of total live cells (g), monocytes (h), macrophages (i),
neutrophils (m), lymphocytes (n), and plasma cells (o) are presented as million cells infiltrated into the
peritoneum. Infiltrating macrophages were further characterized as avacuolar (less than 50% cytoplasm

RI
vacuolar, j) or vacuolar (>50%, k). Cells from the monocyte lineage deemed macrophages were
compared to total (Macrophages/ [monocytes+macrophages], l).

SC
Figure 2. Influence of FO supplementation on cytokine production, phagocytosis, and gene expression
of isolated peritoneal cells. White bars indicate CTL, black bars indicate FO. Results are presented as
means ± SEM, with statistically significant treatment effects denoted by * and significant time effects

U
denoted by letters (2-way ANOVA, p < 0.05). 3% thioglycollate-elicited inflammatory cells (2 days and 4
days) were assessed for ex vivo LPS-stimulated (16 h) cytokine production, namely IL-6 (a), TNFα (b), IL-
AN
10 (c) and IL-1β (d). Recruited inflammatory cells (f) were assessed for ex vivo fluorescent particle
phagocytosis. qPCR was conducted on infiltrated peritoneal exudate cells on day 4 (90-95%
macrophages) to determine mRNA content of proteins involved the classical M1/M2 transition,
inflammatory cell efferocytosis, and fat metabolism (e). Results for mRNA are presented as means ±
M

SEM and significance was determined via t-test with p < 0.05 (*). p-values less than 0.10 are shown.
D

Figure 3. Influence of FO supplementation on content of lipid mediators in peritoneal exudates in


response to chemical peritonitis. White bars indicate CTL, black bars indicate FO. Results are expressed
as means ± SEM, with statistically significant treatment effects denoted by * and Bonferroni post-tests
TE

denoted by letters (2-way ANOVA, p < 0.05). Day 0, 2, and 4 post-thioglycollate injection peritoneal
exudate washings (5 mL) were assessed for concentration of Prostaglandin E2 (a), Leukotriene B4 (b),
and Resolvin D1 (c) via ELISA. Ratios between Resolvin D1 and Prostaglandin E2 content (R/P ratio, d),
EP

Resolvin D1 and Leukotriene B4 content (R/L ratio, e), and Resolvin D1 and the combination of
Prostaglandin E2 and Leukotriene B4 content (R/[P+L] ratio, f) were calculated.

Table 1. Significant findings are indicated by an arrow (p < 0.05). No change is indicated by a (-).
C

Parameters displayed include baseline weight of animals (initiation of AIN 93 diet, in g), food intake (g)
animal weight change (at date of euthanization, in g), immune cell populations in the peritoneum at the
AC

time of sacrifice (million cells), ex vivo cytokine production in response to LPS (pg/mL), mRNA expression
of macrophage differentiation markers in cells harvested on day 4 (90-95% macrophages, relative),
bioparticle phagocytosis of harvested peritoneal cells (relative), and lipid mediators (Prostaglandin E2,
Leukotriene B4, and Resolvin D1) found in the exudate (pg/mL).

Table 1. Summary of effects of fish oil supplementation on biochemical parameters in response to


chemical peritonitis.

Treatment effect of FO
ACCEPTED MANUSCRIPT
16

Objective Animal Data


Animal Weight -
Food Intake -
Weight Change -

Peritoneal Cell Counts

PT
Total Live Cells -
Monocytes
Macrophages -

RI
Macrophages/(Monocytes+Macrophages)
Neutrophils -
Lymphocytes -

SC
Plasma Cells -

ex vivo cytokine production

U
IL-6 -
IL-10 -
AN
TNFα
IL-1β -
M

Macrophage gene expression


Arg1 -
CD36 -
D

IL-6 -
iNOS -
TE

PPARγ
Ym2 -
12/15 LO -
EP

ex vivo bioparticle phagocytosis -


C

Peritoneal exudate lipid mediators


PGE2 -
AC

LTB4
RvD1 -

RvD1/PGE2
RvD1/LTB4
RvD1/(PGE2+LTB4)
ACCEPTED MANUSCRIPT
17

References

1 Serhan, C. N. et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation
circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med 196,
1025-1037., doi:10.1084/jem.20020760 (2002).
2 Serhan, C. N. et al. Maresins: novel macrophage mediators with potent antiinflammatory and
proresolving actions. J Exp Med 206, 15-23, doi:10.1084/jem.20081880 (2009).

PT
3 Spite, M. et al. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis.
Nature 461, 1287-1291, doi:10.1038/nature08541 (2009).
4 Serhan, C. N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 510, 92-
101, doi:10.1038/nature13479 (2014).

RI
5 Recchiuti, A. et al. Immunoresolving actions of oral resolvin D1 include selective regulation of
the transcription machinery in resolution-phase mouse macrophages. FASEB J 28, 3090-3102,
doi:10.1096/fj.13-248393 (2014).

SC
6 Titos, E. et al. Resolvin D1 and its precursor docosahexaenoic acid promote resolution of adipose
tissue inflammation by eliciting macrophage polarization toward an M2-like phenotype. J
Immunol 187, 5408-5418, doi:10.4049/jimmunol.1100225 (2011).

U
7 Mayer, K. & Seeger, W. Fish oil in critical illness. Curr Opin Clin Nutr Metab Care 11, 121-127,
doi:10.1097/MCO.0b013e3282f4cdc6 (2008).
AN
8 Sijben, J. W. & Calder, P. C. Differential immunomodulation with long-chain n-3 PUFA in health
and chronic disease. Proc Nutr Soc 66, 237-259, doi:10.1017/S0029665107005472 (2007).
9 Serhan, C. N. Systems approach to inflammation resolution: identification of novel anti-
inflammatory and pro-resolving mediators. J Thromb Haemost 7, 44-48, doi:10.1111/j.1538-
M

7836.2009.03396.x (2009).
10 Grimm, H. et al. Improved fatty acid and leukotriene pattern with a novel lipid emulsion in
surgical patients. Eur J Nutr 45, 55-60, doi:10.1007/s00394-005-0573-8 (2006).
D

11 Dickinson, A., Blatman, J., El-Dash, N. & Franco, J. C. Consumer usage and reasons for using
dietary supplements: report of a series of surveys. J Am Coll Nutr 33, 176-182. doi:
110.1080/07315724.07312013.07875423. (2014).
TE

12 Braga, M. Perioperative immunonutrition and gut function. Curr Opin Clin Nutr Metab Care 15,
485-488, doi:10.1097/MCO.0b013e3283567d8f (2012).
13 Song, G. M. et al. Immunonutrition Support for Patients Undergoing Surgery for Gastrointestinal
EP

Malignancy: Preoperative, Postoperative, or Perioperative? A Bayesian Network Meta-Analysis


of Randomized Controlled Trials. Medicine (Baltimore) 94, e1225,
doi:10.1097/MD.0000000000001225 (2015).
14 Heller, A. R. et al. Omega-3 fatty acids improve the diagnosis-related clinical outcome. Crit Care
C

Med 34, 972-979, doi:10.1097/01.CCM.0000206309.83570.45 (2006).


15 Tsekos, E., Reuter, C., Stehle, P. & Boeden, G. Perioperative administration of parenteral fish oil
AC

supplements in a routine clinical setting improves patient outcome after major abdominal
surgery. Clin Nutr 23, 325-330 (2004).
16 Yoshihara, T. et al. Omega 3 Polyunsaturated Fatty Acids Suppress the Development of Aortic
Aneurysms Through the Inhibition of Macrophage-Mediated Inflammation. Circ J 79, 1470-1478,
doi:10.1253/circj.CJ- 14-0471 (2015).
17 Jung, U. J. et al. Fatty acids regulate endothelial lipase and inflammatory markers in
macrophages and in mouse aorta: a role for PPARgamma. Arterioscler Thromb Vasc Biol 32,
2929-2937, doi:10.1161/ATVBAHA.112.300188. (2012).
ACCEPTED MANUSCRIPT
18

18 Tomasdottir, V., Vikingsson, A., Freysdottir, J. & Hardardottir, I. Dietary fish oil reduces the acute
inflammatory response and enhances resolution of antigen-induced peritonitis. J Nutr Biochem
24, 1758-1765, doi:10.1016/j.jnutbio.2013.03.005 (2013).
19 Wehner, S. et al. Preoperative short-term parenteral administration of polyunsaturated fatty
acids ameliorates intestinal inflammation and postoperative ileus in rodents. Langenbeck's
archives of surgery / Deutsche Gesellschaft fur Chirurgie 397, 307-315, doi:10.1007/s00423-011-
0862-z (2012).

PT
20 Li, S. et al. Defective phagocytosis of apoptotic cells by macrophages in atherosclerotic lesions of
ob/ob mice and reversal by fish oil diet. Circ Res 105, 1072-1082,
doi:10.1161/CIRCRESAHA.109.199570 (2009).

RI
21 Sorensen, L. S. et al. Effects of perioperative supplementation with omega-3 fatty acids on
leukotriene B4 and leukotriene B5 production by stimulates neutrophils in patients with
colorectal cancer: a randomized, placebo-controlled intervention trial. Nutrients 6, 4043-4057,
doi:10.3390/nu6104043 (2014).

SC
22 Sorensen, L. S. et al. Randomized clinical trial of perioperative omega-3 fatty acid supplements
in elective colorectal cancer surgery. Br J Surg 101, 33-42, doi:10.1002/bjs.9361 (2014).
23 Jellema, A., Plat, J. & Mensink, R. P. Weight reduction, but not a moderate intake of fish oil,

U
lowers concentrations of inflammatory markers and PAI-1 antigen in obese men during the
fasting and postprandial state. European journal of clinical investigation 34, 766-773,
AN
doi:10.1111/j.1365-2362.2004.01414.x (2004).
24 Kratz, M. et al. n3 PUFAs do not affect adipose tissue inflammation in overweight to moderately
obese men and women. J Nutr 143, 1340-1347, doi:10.3945/jn.113.174383 (2013).
25 Ma, C. J. et al. Prospective double-blind randomized study on the efficacy and safety of an n-3
M

fatty acid enriched intravenous fat emulsion in postsurfical gastric and colorectal cancer
patients. Nutri J 14, 9, doi:10.1186/1475-2891-14-9 (2015).
26 Schneider, M. Collecting resident or thioglycollate-elicited peritoneal macrophages. Methods
D

Mol Biol 1031, 37-40, doi:10.1007/978-1-62703-481-4_4 (2013).


27 Phillips, B. E. et al. Impaired recovery from peritoneal inflammation in a mouse model of mild
dietary zinc restriction. Mol Nutr Food Res, doi:10.1002/mnfr.20150068 (2015).
TE

28 Rossi, A. et al. In vivo sex differences in leukotriene biosynthesis in zymosan-induced peritonitis.


Pharmacological research 87, 1-7, doi:10.1016/j.phrs.2014.05.011 (2014).
29 Yoon, Y. S. et al. PPARγ activation following apoptotic cell instillation promotes resolution of
EP

lung inflammation and fibrosis via regulation of efferocytosis and proresolving cytokines.
Mucosal Immunol 8, 1031-1046, doi:10.1038/mi.2014.130 (2015).
30 Davis, M. J. et al. Macrophage M1/M2 polarization dynamically adapts to changes in cytokine
microenvironments in Cryptococcus neoformans infection. Mbio 4, e00264-00213,
C

doi:10.1128/mBio.00264-13 (2013).
31 Chang, H. Y., Lee, H. N., Kim, W. & Surh, Y. J. Docosahexaenoic acid induces M2 macrophage
AC

polarization through peroxisome proliferator-activated receptor gamma activation. Life Sci 120,
39-47 doi:10.1016/j.lfs.2014.10.014 (2015).
32 Green-Mitchell, S. M. et al. Deletion of 12/15-lipoxygenase alters macrophage and islet function
in NOD-Alox15(null) mice, leading to protection against type 1 diabetes development. PLoS One
8, e56763, doi:10.1371/journal.pone.0056763 (2013).
33 Folch, J., Lees, M. & Sloane Stanley, G. H. A simple method for the isolation and purification of
total lipides from animal tissues. J Biol Chem 226, 497-509 (1957).
34 Chiu, C. Y. et al. Omega-6 docosapentaenoic acid-derived resolvins and 17-
hydroxydocosahexaenoic acid modulate macrophage function and alleviate experimental colitis.
Inflamm Res 61, 967-976, doi:10.1007/s00011-012-0489-8 (2012).
ACCEPTED MANUSCRIPT
19

35 Orr, S. K., Colas, R. A., Dalli, J., Chiang, N. & Serhan, C. N. Proresolving actions of a new resolvin
D1 analog mimetic qualifies as an immunoresolvent. Am J Physiol Lung Cell Mol Physiol 308,
L904-L911, doi:10.1152/ajplung.00370.2014 (2015).
36 Harris, W. S. Expert opinion: omega-3 fatty acids and bleeding-cause for concern? Am J Cardiol
99, 44C-46C, doi:doi:10.1016/j.amjcard.2006.11.021 (2007).
37 Wales, K. M. et al. N-3 PUFAs protect against aortic inflammation and oxidative stress in
angiotensin II-infused apolipoprotein E-/- mice. PLoS One. 9, e112816. doi:

PT
112810.111371/journal.pone.0112816. eCollection 0112014. (2014).
38 Poulsen, R. C., Gotlinger, K. H., Serhan, C. N. & Kruger, M. C. Identification of inflammatory and
proresolving lipid mediators in bone marrow and their lipidomic profiles with ovariectomy and

RI
omega-3 intake. Am J Hematol. 83, 437-445. doi: 410.1002/ajh.21170. (2008).
39 Isobe, Y. et al. Identification and structure determination of novel anti-inflammatory mediator
resolvin E3, 17,18-dihydroxyeicosapentaenoic acid. J Biol Chem 287, 10525-10534,
doi:10.1074/jbc.M112.340612 (2012).

SC
40 Arnardottir, H. H., Dalli, J., Colas, R. A., Shinohara, M. & Serhan, C. N. Aging delays resolution of
acute inflammation in mice: reprogramming the host response with novel nano-proresolving
medicines. J Immunol 193, 4235-4244, doi:10.4049/jimmunol.1401313 (2014).

U
41 Dalli, J. & Serhan, C. N. Specific lipid mediator signatures of human phagocytes: microparticles
stimulate macrophage efferocytosis and pro-resolving mediators. Blood 120, e60-e72,
AN
doi:10.1182/blood-2012-04-423525 (2012).
42 Lee, H. N., Kundu, J. K., Cha, Y. N. & Surh, Y. J. Resolvin D1 stimulates efferocytosis through
p50/p50-mediated suppression of tumor necrosis factor-alpha expression. J Cell Sci 126, 4037-
4047, doi:10.1242/jcs.131003 (2013).
M

43 Schif-Zuck, S. et al. Saturated-efferocytosis generates pro-resolving CD11b low macrophages:


modulation by resolvins and glucocorticoids. Eur J Immunol 41, 366-379,
doi:10.1002/eji.201040801 (2011).
D

44 Schwab, J. M., Chiang, N., Arita, M. & Serhan, C. N. Resolvin E1 and protectin D1 activate
inflammation-resolution programmes. Nature 447, 869-874, doi:10.1038/nature05877 (2007).
45 Serhan, C. N. et al. Macrophage proresolving mediator maresin 1 stimulates tissue regeneration
TE

and controls pain. FASEB J 26, 1755-1765, doi:10.1096/fj.11-201442 (2012).


46 Arita, M. et al. Metabolic inactivation of resolvin E1 and stabilization of its anti-inflammatory
actions. J Biol Chem 281, 22847-22854, doi:10.1074/jbc.M603766200 (2006).
EP

47 Babaahmadi Rezaei, H., Doosti, M., Aminian, M. & Shabani, P. Compare the effect of
eicosapentaenoic acid and oxidized low-density lipoprotein on the expression of CD36 and
peroxisome proliferator-activated receptor gamma. Iran Biomed J. 17, 84-92.,
doi:10.6091/ibj.11322.2013 (2013).
C

48 Finstad, H. S. et al. Cell proliferation, apoptosis and accumulation of lipid droplets in U937-1 cells
incubated with eicosapentaenoic acid. Biochem J 336, 451-459, doi:10.1042/bj3360451 (1998).
AC

49 McLaren, J. E., Michael, D. R., Guschina, I. A., Harwood, J. L. & Ramji, D. P. Eicosapentaenoic acid
and docosahexaenoic acid regulate modified LDL uptake and macropinocytosis in human
macrophages. Lipids 46, 1053-1061, doi:0.1007/s11745-011-3598-1 (2011).
50 Jiang, C., Ting, A. T. & Seed, B. PPAR-gamma agonists inhibit production of monocyte
inflammatory cytokines. Nature 391, 82-86, doi:10.1038/34184 (1998).
51 Dalli, J. et al. The novel 13S,14S-epoxy-maresin is converted by human macrophages to maresin
1 (MaR1), inhibits leukotriene A4 hydrolase (LTA4H), and shifts macrophage phenotype. FASEB J
27, 2573-2583, doi:10.1096/fj.13-227728 (2013).
52 Arita, M. et al. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23
to regulate inflammation. J Immunol 178, 3912-3917, doi:10.4049/jimmunol.178.6.3912 (2007).
ACCEPTED MANUSCRIPT
20

53 Scott, M. J. et al. Leukotriene B4 receptor (BLT-1) modulates neutrophil influx into the
peritoneum but not the lung and liver during surgically induced bacterial peritonitis in mice. Clin
Diagn Lab Immunol 11, 936-941., doi:10.1128/CDLI.11.5.936-941.2004 (2004).
54 Mayer, K. et al. Parenteral nutrition with fish oil modulates cytokine response in patients with
sepsis. Am J Respir Crit Care Med. 167, 1321-1328. Epub 2003 Feb 1325. (2003).
55 Senkal, M. et al. Supplementation of omega-3 fatty acids in parenteral nutrition beneficially
alters phospholipid fatty acid pattern. JPEN J Parenter Enteral Nutr 21, 12-17,

PT
doi:10.1177/014860710703100112 (2007).
56 Ryan, A., Power, D. G. & Reynolds, J. V. Immuno-nutrition in upper gastrointestinal surgery. Ann
Surg 249, 1062-1063, doi:10.1097/SLA.0b013e3181a8305d (2009).

RI
U SC
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT

PT
a) b) c)

RI
U SC
d) e) f)

Live Cells
AN
Monocytes Macrophages
Cell Number

40
Cell Number

Cell Number
15 20
b
(1x10^6)

(1x10^6)

(1x10^6)
M
30 15
10
20 a 10
5 *
10 a* 5
*
0
D

0 0
0

4
g) Day h) Day i) Day
TE

Avacuolar Vacuolar Monocyte Lineage


Macrophages Macrophages Maturity
Macrophages)
Macrophages/
(Monocytes +
Cell Number

Cell Number

20 10 1.5
(1x10^6)

(1x10^6)
EP

15 8 *
6 1.0 *
10 *
4 0.5
5 2
0 0 0.0
C
0

4
0

j) Day k) Day l) Day


AC

Neutrophils Lymphocytes Plasma Cells


Cell Number
Cell Number

Cell Number

6 2.0 0.8
(1x10^6)
(1x10^6)

(1x10^6)

1.5 0.6
4
1.0 0.4
2
0.5 0.2
0 0.0 0.0
0

4
0

m) Day n) Day o) Day


ACCEPTED MANUSCRIPT

Macrophage mRNA Profile

p = 0.10
p = 0.003
1.5
α

Relative mRNA
IL-6 TNF

content
1000 2500 * 1.0 *
b

pg/mL

PT
800
pg/mL

2000
600 1500 0.5
400 1000 a *
200 500
0 0 0.0

RI
2

12 Y γ
2

4
a) Day b) e)

1
36

P OS
Day

/1 m2
iN -6

LO
R
rg

IL

A
D

5
A
C
SC
IL-10 IL-1 β Macrophage

Phagocytosis
80 a 200 1.5

Relative
pg/mL
pg/mL

60 150 1.0

U
40 100
b 0.5
20 50
0
0
Day
AN 0.0
2

4
2

4
c) d) Day f) Day
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT

PT
RI
PGE2 R/P ratio
40 6
RvD1/PGE2
*

SC
30 *
pg/mL

4 *
20
2
10
0 0

U
0

4
0

a) Day d) Day

LTB4
AN
R/L ratio
80 b 6 *
RvD1/LTB4

M
60
pg/mL

4
40 a * a *
*
a* b b*
2
20
D

0 0
0

4
0

b) Day e) Day
TE
RvD1/(PGE2+LTB4)

RvD1 R/(P+L) ratio


100 3 *
80
EP
pg/mL

60 2 a *
40 b b*
1
20
0 0
C
0

c) Day f) Day
AC

You might also like