a l eo f c y tokin r n iol u o i gy journal of cytokine ... · pdf fileand a cocktail of...

5
Unexpected Macrophage Activity Alters the Inflammatory Process in Necrotizing Enterocolitis Jessica L. Raque 1,2* , Paul J. Matheson 1,2 , Kevin Connolly 1,2 , Jessica A. Shepherd 1,2 , Eric A. Stamper 1,2 , Kelly M. McMasters 1,2 , Victoria Graham 1,2 , R. Neal Garrison 1,2 , Jason W. Smith 1,2 and Cynthia D. Downard 1,2 1 Hiram C. Polk, Jr., M.D. Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA 2 Robley Rex Veterans Affairs Hospital, Louisville, KY 40206, USA * Corresponding author: Jessica L. Raque, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA, Tel: 502-629-8630; Fax: 502-852-8915; E-mail: [email protected] Received date: May 13, 2017, Accepted date: May 31, 2017, Published date: June 06, 2017 Copyright: © 2017 Raque Jl, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Abstract Objective: Macrophages play a role in clearing bacteria from the gut in the initial stages of necrotizing enterocolitis (NEC). Macrophage Inflammatory Protein-1α (MIP-1α), also known as CCL3, is a chemokine produced by macrophages that enhances the immune response. It acts as both a recruiter of immune cells and a coactivator of other macrophages. Interleukin-12 (IL-12) is a pro-inflammatory chemotactant produced by several types of phagocytic cells such as dendritic cells and neutrophils, but primarily by macrophages. We hypothesized that the pro-inflammatory state associated with NEC pathophysiology would induce increased expression of MIP-1α and IL-12 that would be detectable in serum and intestinal tissue. Methods: Timed pregnant Sprague-Dawley rats were randomized by litter. Controls were delivered vaginally and dam-fed. NEC pup groups were delivered 12 h prematurely via Cesarean section, formula fed, given a single oral dose of lipopolysaccharide, and subjected to intermittent cold and hypoxia as part of a proven NEC rat model protocol. Animals were sacrificed at 0, 12, 24, 48, 72, and 96 h of life and serum and intestinal tissue samples were collected. Samples were analysed via western blot using antibodies with affinity to MIP-1α and IL-12 respectively. Results: Serum and ileal levels of MIP-1 alpha were increased from 48 to 72 h in NEC animals when compared with controls. Serum and ileal IL-12 was downregulated in NEC groups at 12 and 24 h compared to controls. Conclusion: Macrophage function plays an important role in the first 48 h of NEC pathophysiology. Downregulation of IL-12 in the setting of increased MIP-1α expression may represent deranged or inhibited macrophage function in the context of NEC pathogenesis. Further work to elucidate the significance of these findings is warranted. Keywords: Necrotizing enterocolitis; Macrophage; Cytokine; Chemokine; MIP-1 alpha; CCL3; IL-12 Introduction Necrotizing enterocolitis (NEC) is a devastating gastrointestinal illness primarily affecting premature infants. NEC is characterized by epithelial destruction leading to gut barrier failure, and requires surgery in 20-40% of those affected. Of infants requiring operative intervention, 20-50% will die from their disease [1-3]. Despite years of research into the cause, prevention, and treatment of NEC, the mortality rate has remained the same over the last three decades, and no definitive cause has been found [4,5]. Macrophages have been shown to play an important role in the clearance of bacteria in the initial stages of NEC [6]. Activation of macrophages is characterized by the release of many intercellular mediators including cytokines, growth factors, and chemokines. Macrophage inflammatory protein 1-alpha (MIP-1α) (also known as CCL3) is a chemokine that is upregulated in macrophages in response to stressors such as exposure to bacterial lipopolysaccharide (LPS) [7,8]. Interleukin-12 (IL-12) is a pro-inflammatory cytokine produced by macrophages, dendritic cells, and B cells, and works to activate macrophages and recruit phagocytic cells to sites of infection. Mice that are deficient in IL-12 or its receptor are more susceptible to bacterial infections than their normal counterparts [9-11]. We hypothesized that the pro-inflammatory state associated with NEC would lead to an up-regulation of both MIP-1α and IL-12. Materials and Methods Approval for the study was granted by the Institutional Animal Care and Use Committee (IACUC), the Biohazard Safety Committee, and the Research and Development Committee of the Robley Rex Veteran’s Affairs Hospital in Louisville, KY. Timed pregnant Sprague-Dawley rats (Harlan, Indianapolis, IN) were housed for one week prior to initiation of experimental protocol, and allowed water and rat chow ad lib. Dams were then randomized to the Control or NEC group and their pups were assigned accordingly aſter birth. Control dams delivered their pups vaginally. Control pups were kept with their mothers and allowed to nurse on demand until the time of sacrifice for tissue and Raque et al., J Cytokine Biol 2017, 2:2 Research Article OMICS International Volume 2 • Issue 2 • 1000115 Journal of Cytokine Biology DOI: 10.4172/2576-3881.1000115 J Cytokine Biol, an open access journal ISSN:2576-3881 J o u r n a l o f C yt o k i n e B i o l o g y ISSN: 2576-3881

Upload: halien

Post on 28-Mar-2018

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: a l eo f C y tokin r n iol u o i gy Journal of Cytokine ... · PDF fileand a cocktail of phosphatase (P5726, Sigma-Aldrich) and protease inhibitors (P8340, Sigma-Aldrich), and protein

Unexpected Macrophage Activity Alters the Inflammatory Process inNecrotizing EnterocolitisJessica L. Raque1,2*, Paul J. Matheson1,2, Kevin Connolly1,2, Jessica A. Shepherd1,2, Eric A. Stamper1,2, Kelly M. McMasters1,2, Victoria Graham1,2, R. NealGarrison1,2, Jason W. Smith1,2 and Cynthia D. Downard1,2

1Hiram C. Polk, Jr., M.D. Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA2Robley Rex Veterans Affairs Hospital, Louisville, KY 40206, USA*Corresponding author: Jessica L. Raque, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA, Tel: 502-629-8630; Fax:502-852-8915; E-mail: [email protected]

Received date: May 13, 2017, Accepted date: May 31, 2017, Published date: June 06, 2017

Copyright: © 2017 Raque Jl, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricteduse, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Objective: Macrophages play a role in clearing bacteria from the gut in the initial stages of necrotizingenterocolitis (NEC). Macrophage Inflammatory Protein-1α (MIP-1α), also known as CCL3, is a chemokine producedby macrophages that enhances the immune response. It acts as both a recruiter of immune cells and a coactivatorof other macrophages. Interleukin-12 (IL-12) is a pro-inflammatory chemotactant produced by several types ofphagocytic cells such as dendritic cells and neutrophils, but primarily by macrophages. We hypothesized that thepro-inflammatory state associated with NEC pathophysiology would induce increased expression of MIP-1α andIL-12 that would be detectable in serum and intestinal tissue.

Methods: Timed pregnant Sprague-Dawley rats were randomized by litter. Controls were delivered vaginally anddam-fed. NEC pup groups were delivered 12 h prematurely via Cesarean section, formula fed, given a single oraldose of lipopolysaccharide, and subjected to intermittent cold and hypoxia as part of a proven NEC rat modelprotocol. Animals were sacrificed at 0, 12, 24, 48, 72, and 96 h of life and serum and intestinal tissue samples werecollected. Samples were analysed via western blot using antibodies with affinity to MIP-1α and IL-12 respectively.

Results: Serum and ileal levels of MIP-1 alpha were increased from 48 to 72 h in NEC animals when comparedwith controls. Serum and ileal IL-12 was downregulated in NEC groups at 12 and 24 h compared to controls.

Conclusion: Macrophage function plays an important role in the first 48 h of NEC pathophysiology.Downregulation of IL-12 in the setting of increased MIP-1α expression may represent deranged or inhibitedmacrophage function in the context of NEC pathogenesis. Further work to elucidate the significance of thesefindings is warranted.

Keywords: Necrotizing enterocolitis; Macrophage; Cytokine;Chemokine; MIP-1 alpha; CCL3; IL-12

IntroductionNecrotizing enterocolitis (NEC) is a devastating gastrointestinal

illness primarily affecting premature infants. NEC is characterized byepithelial destruction leading to gut barrier failure, and requiressurgery in 20-40% of those affected. Of infants requiring operativeintervention, 20-50% will die from their disease [1-3]. Despite years ofresearch into the cause, prevention, and treatment of NEC, themortality rate has remained the same over the last three decades, andno definitive cause has been found [4,5].

Macrophages have been shown to play an important role in theclearance of bacteria in the initial stages of NEC [6]. Activation ofmacrophages is characterized by the release of many intercellularmediators including cytokines, growth factors, and chemokines.Macrophage inflammatory protein 1-alpha (MIP-1α) (also known asCCL3) is a chemokine that is upregulated in macrophages in responseto stressors such as exposure to bacterial lipopolysaccharide (LPS)[7,8].

Interleukin-12 (IL-12) is a pro-inflammatory cytokine produced bymacrophages, dendritic cells, and B cells, and works to activatemacrophages and recruit phagocytic cells to sites of infection. Micethat are deficient in IL-12 or its receptor are more susceptible tobacterial infections than their normal counterparts [9-11].

We hypothesized that the pro-inflammatory state associated withNEC would lead to an up-regulation of both MIP-1α and IL-12.

Materials and MethodsApproval for the study was granted by the Institutional Animal Care

and Use Committee (IACUC), the Biohazard Safety Committee, andthe Research and Development Committee of the Robley Rex Veteran’sAffairs Hospital in Louisville, KY. Timed pregnant Sprague-Dawleyrats (Harlan, Indianapolis, IN) were housed for one week prior toinitiation of experimental protocol, and allowed water and rat chow adlib.

Dams were then randomized to the Control or NEC group and theirpups were assigned accordingly after birth. Control dams deliveredtheir pups vaginally. Control pups were kept with their mothers andallowed to nurse on demand until the time of sacrifice for tissue and

Raque et al., J Cytokine Biol 2017, 2:2

Research Article OMICS International

J Cytokine Biol, an open access journal

Volume 2 • Issue 2 • 1000115

Jour

nal of Cytokine Biology

Journal of Cytokine BiologyDOI: 10.4172/2576-3881.1000115

J Cytokine Biol, an open access journalISSN:2576-3881

Jour

nal o

f Cytokine Biology

ISSN: 2576-3881

Page 2: a l eo f C y tokin r n iol u o i gy Journal of Cytokine ... · PDF fileand a cocktail of phosphatase (P5726, Sigma-Aldrich) and protease inhibitors (P8340, Sigma-Aldrich), and protein

serum harvest. Time matched NEC dams underwent carbon dioxideanesthesia and decerebration prior to rapid cesarean delivery of theirpups 12 h prematurely. NEC pups were revived and kept in a warmedincubator at 37.0 degrees centigrade.

A previously published protocol was used to induce NEC in theNEC pups [12,13]. Briefly, the pups were orally gavage fed formulaconsisting of 20 g Similac 60/40 (Ross Pediatrics, Columbus, OH)dissolved in 100 mL Esbilac puppy formula (Pet-Ag, New Hampshire,IL) every four hours and were given one oral dose oflipopolysaccharide (2 mg/kg Salmonella enterica serotypetyphimurium; Sigma Chemical CO., St. Louis, MO) at 12 h of life.Feeds were started at 0.1 mL/feed at time 0 and advanced to 0.4 mL/feed at 96 h for approximately 836.8 kJ/kg/day. Feeds and the oral doseof LPS were administered via a silastic 1.9 French single lumen catheter(Utah Medical Products, Inc., Midvale, UT) gently advanced into thestomach. The position was verified by the appearance of formula in thestomach during instillation of formula. NEC pups were also subjectedto hypoxia (100% nitrogen gas for 60 sec) and hypothermia (4°C forten minutes) every 12 h.

Control and NEC pups were sacrificed at 0, 12, 24, 48, 72, and 96 hof life. Both Control and NEC ileum samples were homogenized inRIPA lysis buffer (R0278 Sigma-Aldrich Chemical Co., St. Louis, MO)and a cocktail of phosphatase (P5726, Sigma-Aldrich) and proteaseinhibitors (P8340, Sigma-Aldrich), and protein concentrations weredetermined. Samples containing equal amounts of 35 μg of totalprotein were resolved by SDS-PAGE under reducing conditions. Afterelectrophoresis, proteins were transferred to a polyvinylidenedifluoride membrane and probed with antibodies against CCL-3 andIL-12 with a 1:1000 dilution in BSA. Each protein sample wasstandardized to its own β-actin level.

Ileum sections for Control and NEC groups from the 48 h timepoint were mounted in paraffin and stained with either H&E orImmunohistochemistry (IHC). The following scale was used to gradeeach sample: grade 0, normal and no damage; grade 1, epithelial celllifting or separation; grade 2, sloughing of epithelial cells to mid villuslevel; grade 3, necrosis of the entire villus; and grade 4, transmuralnecrosis [14]. These samples were selected for IHC based on our priorstudies showing a significant difference in intestinal blood flow at thisplace in the NEC protocol timeline [15]. IHC staining was performedusing rabbit anti-rodent polyclonal antibodies for MIP-1α (CCL-3) andIL-12. The slides for IHC were graded into one of four differentcategories: (-) no significant staining, (+) slight positive, (++)moderately positive, and (+++) strongly positive.

Serum samples were taken at the time of sacrifice and prepared foranalysis per the manufacturer’s instructions using a Luminex MagPixmultianalyte cytokine array (Luminex, 23-plex cytokine panel specificfor rat). Two-way analysis of variance (ANOVA) using SigmaPlot forWindows 11.1.0.102 (Systat Software, Inc., San Jose, CA) was used todetermine the difference between groups and time points, and when adifference was found, it was confirmed with the Tukey-Kramerhonestly significant difference test. The null hypothesis was rejected atp<0.05.

ResultsHistological signs of NEC became apparent in the NEC groups at 24

h, and were most pronounced at 48 h (Figure 1). All animals in theNEC group had at least Grade 1 histological signs of NEC by 24 h, andthe majority of animals had Grade 2 changes by 48 h (Figure 2). All

animals in the Control group maintained Grade 0 histologythroughout the course of the study.

Figure 1.Histogical diffrences became apparent and most pronounced at the48 h time point.

Figure 2. Histopathological grading of individual animals Controland NEC. Majority of NEC animals had Grade 2 changes by the 48h time point. All animals in the Control group maintained aGrade of 0 throughout the course of the study.

Serum levels of MIP-1α at 0, 12, 24, 48, 72, and 96 h are shown inFigure 3. There was no difference in the amount of MIP-1α present inNEC versus Control animals at the 0, 12, or 24 h time points. The levelof MIP-1α started off somewhat high in both NEC and Controlanimals and experienced a significant drop between 12 and 24 h in

Citation: Raque JL, Matheson PJ, Connolly K, Shepherd JA, Stamper EA, et al. (2017) Unexpected Macrophage Activity Alters the InflammatoryProcess in Necrotizing Enterocolitis. J Cytokine Biol 2: 115.

Page 2 of 5

J Cytokine Biol, an open access journalVolume 2 • Issue 2 • 1000115

Histopathological grading of Control and NEC animals.

DOI: 10.4172/2576-3881.1000115

J Cytokine Biol, an open access journalISSN:2576-3881

doi:10.4172/2576-3881.1000115

Page 3: a l eo f C y tokin r n iol u o i gy Journal of Cytokine ... · PDF fileand a cocktail of phosphatase (P5726, Sigma-Aldrich) and protease inhibitors (P8340, Sigma-Aldrich), and protein

both groups. After 24 h, the Control animals continued to experience adecrease in MIP-1α, whereas the NEC animals showed elevated levels.

After an initial drop, Interleukin-12 levels were steady in NECanimals throughout the NEC protocol (Figure 4). Levels of IL-12 weresignificantly lower in NEC animals when compared to controls at 12and 24 h. Following this, the Control animals experienced a drop inIL-12 while the NEC animals remained unchanged.

Figure 3. There is no difference in MIP-1α levels between the 48 and96 h time points. Conversly, the Control groups decrease at thesetime points.

Figure 4. significantly lower in theNEC groups at the 12 and 24 h time points when compared to theControl. The Controls groups experienced a decrease in the IL-12levels after the 24 h time point.

Immunohistochemistry of Control and NEC groups at the 48 h timepoint revealed similar results to serum levels at the same time point.The Control group (Figure 5A) showed no significant staining forMIP-1α while the NEC group stained strongly positive for MIP-1α(Figure 5B). Similarly for IL-12 the Control group displayed nosignificant staining (Figure 5C). The NEC group at the same time pointstained strongly positive for IL-12 (Figure 5D).

Figure 5. The Control group had no staining for MIP-1α(A). TheNEC group stained strongly positive (B). Images A and B are shownat a 1:100 dilution. IL-12 displayed no staining for the Controlgroup (C) and strongly positive in the NEC group (D). Images Cand D are shown at a 1:100 dilution. All tissues are shown at a 40Xmaginfication.

DiscussionThe exact pathophysiology of NEC continues to be unclear despite

years of research into the cause. Many risk factors have been identifiedas contributing to the pathogenesis of NEC, including prematurity,enteral feeding, hypothermia, bacterial colonization, and hypoxia[16-23]. Many believe NEC to be secondary to a deranged oruncontrolled inflammatory response. Therefore, researchers havesought to elucidate the cascade of inflammatory cytokines found inNEC [24]. As a part of the innate immune system, the macrophageplays an important role in the recognition and clearance of bacteriafrom sites of infection. Present in tissues, macrophages are one ofseveral cell types known as antigen presenting cells (APCs), capable ofrecognizing and engulfing foreign antigens for presentation to otherimmune cells to stimulate the clearance of infection.

Macrophages not only act as APCs and phagocytes, however. Theyalso secrete cytokines, growth factors, and chemokines (cytokines thatare chemoattractants) to recruit additional monocytes (macrophageprecursors), neutrophils, and dendritic cells to the site ofinflammation. Prior work by Emami et al. demonstrated theimportance of macrophages in NEC caused by infection with thebacteria Cronobacter sakazakii [6]. Animals subjected to the bacteriathat were depleted of macrophages and polymorphonuclear leukocytes(PMNs) were found to have higher intestinal bacterial load, higherpathologic scores for intestinal damage, and earlier mortality thanthose with intact macrophages and PMNs.

Citation: Raque JL, Matheson PJ, Connolly K, Shepherd JA, Stamper EA, et al. (2017) Unexpected Macrophage Activity Alters the InflammatoryProcess in Necrotizing Enterocolitis. J Cytokine Biol 2: 115.

Page 3 of 5

J Cytokine Biol, an open access journal

Volume 2 • Issue 2 • 1000115

IL-12 concentration levels were

DOI: 10.4172/2576-3881.1000115

J Cytokine Biol, an open access journalISSN:2576-3881

doi:10.4172/2576-3881.1000115

Page 4: a l eo f C y tokin r n iol u o i gy Journal of Cytokine ... · PDF fileand a cocktail of phosphatase (P5726, Sigma-Aldrich) and protease inhibitors (P8340, Sigma-Aldrich), and protein

Macrophage inflammatory proteins-1α, 1β, and 2 (MIPs) areheparin binding proteins with inflammatory and immunoregulatoryactivity [8]. The MIPs were initially characterized as proteins secretedfrom endotoxin-stimulated mouse macrophages, but further researchdemonstrated that neutrophils, fibroblasts, epithelial cells, and T cellsare also MIP-secreting cells. The three MIPs have distinctcharacteristics, but share a few overlapping roles. All MIPs actprimarily in a pro-inflammatory nature and are chemotactic, acting torecruit and activate other cells at the sites of inflammation or injury.MIP-1α acts with a broader specificity than MIP-1β in that it attracts Bcells, cytotoxic T cells, and CD4+T cells, whereas MIP-1β workspreferentially on CD4+ T cells [25]. The macrophage is thought to bethe primary source for MIPs, but Sherry et al. demonstrated that Tcells are either a major source of MIPs or that they act to markedlyupregulate the production of MIPs by other cell types [7]. In that study,levels of MIP-1α were measured in mice in vitro after intraperitonealinjection of LPS, and were shown to peak at two hours, then fall toundetectable levels by eight hours.

In our study, there were similar amounts of MIP-1α at the 12 and 24h time points in both the NEC and Control groups, and that level wastrending downward. By 48 h, we saw a significantly different increasein MIP-1α in NEC animals compared to controls. This trend continuedin the NEC animals and remained significant until 96 h-the end-pointof our study. As one of the first responders to sites of infection andinflammation, it seems likely that the macrophage response to theinduction of NEC would be nearly immediate, but we did not find thatto be true. Pathological signs of NEC were first apparent at 24 h inNEC animals, but levels of MIP-1α in those animals did not increasesignificantly until 48 h. Additionally, instead of peaking and thenbecoming undetectable, we show a sustained increase in MIP-1α.

These findings are markedly different than those demonstrated inthe prior work by Sherry et al. [7]. It is possible that the lag timebetween histopathological evidence of the disease compared tosignificantly elevated serum levels of MIP-1α in NEC animalsdemonstrates a slow reaction of macrophages in this NEC model. Adelay in macrophage activation might perpetuate further damage tothe intestines, worsening NEC. The sustained increase in MIP-1α inNEC animals demonstrates the continued inflammatory response asmacrophages are attempting to fight off the intestinal inflammation.

IL-12, produced by macrophages, dendritic cells, and B cells, workssynergistically with IL-18 to induce production of interferon-gamma(IFN-γ) from natural killer and B cells. Together, these pro-inflammatory factors can induce intestinal inflammation in mice [26].Prior studies have demonstrated conflicting results regarding theproduction of IL-12 in rats undergoing a NEC-inducing protocol whencompared to dam fed controls. While Nadler et al. showed decreasedIL-12 mRNA expression in NEC animals [27], Halpern et al. showedan increase in IL-12 mRNA in NEC animals and a correlation betweenthe number of IL-12 positive cells and the degree of tissue destruction[28]. The positive correlation between the number of IL-12 positivecells and the amount of tissue damage present led many to draw theconclusion that IL-12 is involved with the initial stages of NEC andmay contribute to the intestinal necrosis associated with NEC [24].

Our findings (i.e., a relatively steady level of IL-12 in NEC animalscompared to a marked elevation of IL-12 at 12 and 24 h in Controlanimals) suggest that macrophage function in NEC animals may bediminished. One might expect a reduced inflammatory reaction, butthis is not confirmed histologically. There is clear progression ofinflammation and necrosis of the intestine in the NEC animals despite

the decreased amount of IL-12. Therefore, although other studies havedemonstrated the involvement of IL-12 in tissue inflammation in theintestine, our findings of low IL-12 with markedly inflamed intestineindicate that further investigation into the activity of IL-12 in NECanimals is warranted.

ConclusionMacrophages play a central role in the elimination of bacteria from

the gut in the initial stages of NEC. Animals undergoing a NEC-inducing protocol experienced a delayed upregulation of MIP-1α andno upregulation of IL-12. This delay in the upregulation of themacrophage-secreted pro-inflammatory cytokine MIP-1α paired witha lack of response by IL-12 in NEC animals likely contributes to thepathophysiology of NEC. Enteral supplementation of IL-12, as we havepreviously done with Relaxin, may offer a mechanistic explanation ofIL-12 and if this cytokine is contributing positively or negatively to thepathophysiology of NEC.

AcknowledgementThe authors would like to acknowledge the technical support of

Amy J. Matheson and Samuel A. Matheson in the performance of thesestudies. The work is also supported by Kosair Charities and the JamesR. Petersdorf Fund of Norton Healthcare.

References1. Petrosyan M, Guner YS, Williams M, Grishin A, Ford HR (2009) Current

concepts regarding the pathogenesis of necrotizing enterocolitis. PediatrSurg Int 25: 309-318.

2. Blakely ML, Lally KP, McDonald S, Brown RL, Barnhart DC, et al. (2005)Postoperative outcomes of extremely low birth-weight infants withnecrotizing enterocolitis or isolated intestinal perforation: a prospectivecohort study by the NICHD Neonatal Research Network. Ann Surg 241:984-989.

3. Rowe MI, Reblock KK, Kurkchubasche AG, Healey PJ (1994) Necrotizingenterocolitis in the extremely low birth weight infant. J Pediatr Surg 29:987-990.

4. Berrington JE, Hearn RI, Bythell M, Wright C, Embleton ND (2012)Deaths in preterm infants: changing pathology over 2 decades. J Pediatr160: 49-53.

5. Horbar JD, Badger GJ, Carpenter JH, Fanaroff AA, Kilpatrick S, et al.(2002) Trends in mortality and morbidity for very low birth weightinfants, 1991-1999. Pediatrics 110: 143-151.

6. Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV (2012) Role ofneutrophils and macrophages in the pathogenesis of necrotizingenterocolitis caused by Cronobacter sakazakii. J Surg Res 172: 18-28.

7. Sherry B, Espinoza M, Manogue KR, Cerami A (1998) Induction of thechemokine beta peptides, MIP-1 alpha and MIP-1 beta, bylipopolysaccharide is differentially regulated by immunomodulatorycytokines gamma-IFN, IL-10, IL-4, and TGF-beta. Mol Med 4: 648-657.

8. Driscoll KE (1994) Macrophage inflammatory proteins: biology and rolein pulmonary inflammation. Exp Lung Res 20: 473-490.

9. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, et al. (2002) Theadaptive immune system. (4th Edition), Garland Science, New York,USA.

10. Göebel A, Kavanagh E, Lyons A, Saporoschetz IB, Soberg C, et al. (2000)Injury induces deficient interleukin-12 production, but interleukin-12therapy after injury restores resistance to infection. Ann Surg 231:253-261.

11. O'Suilleabhain C, O'Sullivan ST, Kelly JL, Lederer J, Mannick JA, et al.(1996) Interleukin-12 treatment restores normal resistance to bacterialchallenge after burn injury. Surgery 120: 290-296.

Citation: Raque JL, Matheson PJ, Connolly K, Shepherd JA, Stamper EA, et al. (2017) Unexpected Macrophage Activity Alters the InflammatoryProcess in Necrotizing Enterocolitis. J Cytokine Biol 2: 115.

Page 4 of 5

J Cytokine Biol, an open access journal Volume 2 • Issue 2 • 1000115

DOI: 10.4172/2576-3881.1000115

J Cytokine Biol, an open access journalISSN:2576-3881

doi:10.4172/2576-3881.1000115

Page 5: a l eo f C y tokin r n iol u o i gy Journal of Cytokine ... · PDF fileand a cocktail of phosphatase (P5726, Sigma-Aldrich) and protease inhibitors (P8340, Sigma-Aldrich), and protein

12. Barlow B, Santulli TV (1975) Importance of multiple episodes of hypoxiaor cold stress on the development of enterocolitis in an animal model.Surgery 77: 687-690.

13. Barlow B, Santulli TV, Heird WC, Pitt J, Blanc WA, et al. (1974) Anexperimental study of acute neonatal enterocolitis-the importance ofbreast milk. J Pediatr Surg 9: 587-595.

14. Caplan MS, Hedlund E, Adler L, Hsueh W (1994) Role of asphyxia andfeeding in a neonatal rat model of necrotizing enterocolitis. PediatrPathol 14: 1017-1028.

15. Walker SK, Matheson PJ, Schreiner MT, Smith JW, Garrison RN, et al.(2013) Intraperitoneal 1.5% Delflex improves intestinal blood flow innecrotizing enterocolitis. J Surg Res 184: 358-364.

16. Downard CD, Grant SN, Matheson PJ, Guillaume AW, Debski R, et al.(2011) Altered intestinal microcirculation is the critical event in thedevelopment of necrotizing enterocolitis. J Pediatr Surg 46: 1023-1028.

17. Lin PW, Stoll BJ (2006) Necrotising enterocolitis. Lancet 368: 1271-1283.18. Hsueh W, Caplan MS, Qu XW, Tan XD, De Plaen IG, et al. (2003)

Neonatal necrotizing enterocolitis: clinical considerations andpathogenetic concepts. Pediatr Dev Pathol 6: 6-23.

19. Kosloske AM (1994) Epidemiology of necrotizing enterocolitis. ActaPaediatr Suppl 396: 2-7.

20. Neu J (1996) Necrotizing enterocolitis: the search for a unifyingpathogenic theory leading to prevention. Pediatr Clin North Am 43:409-432.

21. Krediet TG, van Lelyveld N, Vijlbrief DC, Brouwers HA, Kramer WL, etal. (2003) Microbiological factors associated with neonatal necrotizingenterocolitis: protective effect of early antibiotic treatment. Actapaediatrica 92: 1180-1182.

22. Kliegman RM (1990) Models of the pathogenesis of necrotizingenterocolitis. J Pediatr 117: S2-5.

23. Martinez-Tallo E, Claure N, Bancalari E (1997) Necrotizing enterocolitisin full-term or near-term infants: risk factors. Biol Neonate 71: 292-298.

24. Markel TA, Crisostomo PR, Wairiuko GM, Pitcher J, Tsai BM, et al.(2006) Cytokines in necrotizing enterocolitis. Shock 25: 329-337.

25. Schall TJ, Bacon K, Camp RD, Kaspari JW, Goeddel DV (1993) Humanmacrophage inflammatory protein alpha (MIP-1 alpha) and MIP-1 betachemokines attract distinct populations of lymphocytes. J Exp Med 177:1821-1826.

26. Chikano S, Sawada K, Shimoyama T, Kashiwamura SI, Sugihara A, et al.(2000) IL-18 and IL-12 induce intestinal inflammation and fatty liver inmice in an IFN-gamma dependent manner. Gut 47: 779-786.

27. Nadler EP, Dickinson E, Knisely A, Zhang XR, Boyle P, et al. (2000)Expression of inducible nitric oxide synthase and interleukin-12 inexperimental necrotizing enterocolitis. J Surg Res 92: 71-77.

28. Halpern MD, Holubec H, Dominguez JA, Williams CS, Meza YG, et al.(2002) Up-regulation of IL-18 and IL-12 in the ileum of neonatal ratswith necrotizing enterocolitis. Pediatr Res 51: 733-739.

Citation: Raque JL, Matheson PJ, Connolly K, Shepherd JA, Stamper EA, et al. (2017) Unexpected Macrophage Activity Alters the InflammatoryProcess in Necrotizing Enterocolitis. J Cytokine Biol 2: 115.

Page 5 of 5

J Cytokine Biol, an open access journal Volume 2 • Issue 2 • 1000115

DOI: 10.4172/2576-3881.1000115

J Cytokine Biol, an open access journalISSN:2576-3881

doi:10.4172/2576-3881.1000115