Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by intestinal inflammation and necrosis. Although studies suggest that NEC is mediated by an underdeveloped and compromised epithelial barrier allowing bacterial exposure to an immature immune system,
1- Sharma R.
- Tepas 3rd, J.J.
- Hudak M.L.
- Mollitt D.L.
- Wludyka P.S.
- Teng R.J.
- Premachandra B.R.
Neonatal gut barrier and multiple organ failure: role of endotoxin and proinflammatory cytokines in sepsis and necrotizing enterocolitis.
, 2- Bergmann K.R.
- Liu S.X.
- Tian R.
- Kushnir A.
- Turner J.R.
- Li H.L.
- Chou P.M.
- Weber C.R.
- De Plaen I.G.
Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis.
the mechanism remains unclear and no specific treatment is currently available. Several studies suggest that monocyte activation plays a role in NEC,
3- MohanKumar K.
- Kaza N.
- Jagadeeswaran R.
- Garzon S.A.
- Bansal A.
- Kurundkar A.R.
- Namachivayam K.
- Remon J.I.
- Bandepalli C.R.
- Feng X.
- Weitkamp J.H.
- Maheshwari A.
Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model.
, 4- He Y.M.
- Li X.
- Perego M.
- Nefedova Y.
- Kossenkov A.V.
- Jensen E.A.
- Kagan V.
- Liu Y.F.
- Fu S.Y.
- Ye Q.J.
- Zhou Y.H.
- Wei L.
- Gabrilovich D.I.
- Zhou J.
Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation.
, 5- Pang Y.
- Du X.
- Xu X.
- Wang M.
- Li Z.
Monocyte activation and inflammation can exacerbate Treg/Th17 imbalance in infants with neonatal necrotizing enterocolitis.
and animal and human NEC tissues display a macrophage-rich infiltrate.
3- MohanKumar K.
- Kaza N.
- Jagadeeswaran R.
- Garzon S.A.
- Bansal A.
- Kurundkar A.R.
- Namachivayam K.
- Remon J.I.
- Bandepalli C.R.
- Feng X.
- Weitkamp J.H.
- Maheshwari A.
Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model.
, 6M1 to M2 macrophage polarization in heparin-binding epidermal growth factor-like growth factor therapy for necrotizing enterocolitis.
Infiltrating macrophages during inflammation typically result from the differentiation of monocytes that have left the bloodstream. However, not much is known about the early formative myeloid cell changes in the intestine during NEC, and whether monocyte activation and differentiation contribute to its pathogenesis remains unknown.
Many myeloid cell functions are mediated via the transcription factor NF-κB. On activation of receptors such as toll-like receptors, the catalytic subunit of the IKK complex, inhibitor of kappaB kinase β (IKKβ), becomes activated, which leads to NF-κB nuclear translocation and transcription of NF-κB–target genes, including cytokines/chemokines, adhesion molecules, and cell surface receptors.
7- DiDonato J.A.
- Hayakawa M.
- Rothwarf D.M.
- Zandi E.
- Karin M.
A cytokine-responsive IkappaB kinase that activates the transcription factor NF-kappaB.
Our laboratory has previously shown that NF-κB is persistently activated in the intestine in a neonatal rat model of NEC
8- De Plaen I.G.
- Liu S.X.
- Tian R.
- Neequaye I.
- May M.J.
- Han X.B.
- Hsueh W.
- Jilling T.
- Lu J.
- Caplan M.S.
Inhibition of nuclear factor-kappaB ameliorates bowel injury and prolongs survival in a neonatal rat model of necrotizing enterocolitis.
and treatment with a specific NF-κB–inhibitory peptide decreased NEC-associated mortality and bowel injury.
8- De Plaen I.G.
- Liu S.X.
- Tian R.
- Neequaye I.
- May M.J.
- Han X.B.
- Hsueh W.
- Jilling T.
- Lu J.
- Caplan M.S.
Inhibition of nuclear factor-kappaB ameliorates bowel injury and prolongs survival in a neonatal rat model of necrotizing enterocolitis.
In a murine NEC model, NF-κB was found to be activated in both intestinal inflammatory cells and epithelial cells (IECs) before acute bowel injury.
9- De Plaen I.G.
- Tan X.D.
- Chang H.
- Qu X.W.
- Liu Q.P.
- Hsueh W.
Intestinal NF-kappaB is activated, mainly as p50 homodimers, by platelet-activating factor.
However, whether NF-κB activation in myeloid cells or epithelial cells plays a role in NEC-like injury remains unknown.
In the murine small intestine at birth, myeloid cells are mostly embryonic macrophages, which originate from the yolk sac or fetal liver.
10- Bain C.C.
- Bravo-Blas A.
- Scott C.L.
- Gomez Perdiguero E.
- Geissmann F.
- Henri S.
- Malissen B.
- Osborne L.C.
- Artis D.
- Mowat A.M.
Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice.
At 2 to 3 weeks of age, as the small intestine matures, macrophages derived from monocytes that originated in the bone marrow begin to replace most embryonic macrophages.
10- Bain C.C.
- Bravo-Blas A.
- Scott C.L.
- Gomez Perdiguero E.
- Geissmann F.
- Henri S.
- Malissen B.
- Osborne L.C.
- Artis D.
- Mowat A.M.
Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice.
Although both intestinal macrophages derived from monocytes and embryonic macrophages can be phenotypically identified by Cx3cr1 expression, these populations are functionally unique. Embryonic macrophages have proliferative potential and play a role in tissue debris removal
11- Stevens B.
- Allen N.J.
- Vazquez L.E.
- Howell G.R.
- Christopherson K.S.
- Nouri N.
- Micheva K.D.
- Mehalow A.K.
- Huberman A.D.
- Stafford B.
- Sher A.
- Litke A.M.
- Lambris J.D.
- Smith S.J.
- John S.W.
- Barres B.A.
The classical complement cascade mediates CNS synapse elimination.
, 12Macrophages are required for cell death and tissue remodeling in the developing mouse eye.
, 13- Varol C.
- Mildner A.
- Jung S.
Macrophages: development and tissue specialization.
and vascular development and remodeling.
14- Leid J.
- Carrelha J.
- Boukarabila H.
- Epelman S.
- Jacobsen S.E.
- Lavine K.J.
Primitive embryonic macrophages are required for coronary development and maturation.
, 15- De Kleer I.
- Willems F.
- Lambrecht B.
- Goriely S.
Ontogeny of myeloid cells.
, 16- DeFalco T.
- Bhattacharya I.
- Williams A.V.
- Sams D.M.
- Capel B.
Yolk-sac-derived macrophages regulate fetal testis vascularization and morphogenesis.
, 17- Fantin A.
- Vieira J.M.
- Gestri G.
- Denti L.
- Schwarz Q.
- Prykhozhij S.
- Peri F.
- Wilson S.W.
- Ruhrberg C.
Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction.
In contrast, intestinal macrophages derived from monocytes are terminally differentiated, nonproliferative, and short lived, needing to be continuously replenished by recruitment and differentiation of blood monocytes. During their differentiation into monocyte-derived macrophages, monocytes up-regulate major histocompatibility complex (MHC)-II, which is sequentially followed by down-regulation of Ly6c and Ccr2 and up-regulation of Cx3cr1.
18- Jakubzick C.
- Gautier E.L.
- Gibbings S.L.
- Sojka D.K.
- Schlitzer A.
- Johnson T.E.
- Ivanov S.
- Duan Q.
- Bala S.
- Condon T.
- van Rooijen N.
- Grainger J.R.
- Belkaid Y.
- Ma'ayan A.
- Riches D.W.
- Yokoyama W.M.
- Ginhoux F.
- Henson P.M.
- Randolph G.J.
Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes.
, 19- Bain C.C.
- Scott C.L.
- Uronen-Hansson H.
- Gudjonsson S.
- Jansson O.
- Grip O.
- Guilliams M.
- Malissen B.
- Agace W.W.
- Mowat A.M.
Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors.
, 20Macrophages in intestinal homeostasis and inflammation.
Intestinal resident macrophages are anti-inflammatory and unresponsive to toll-like receptor stimulation.
19- Bain C.C.
- Scott C.L.
- Uronen-Hansson H.
- Gudjonsson S.
- Jansson O.
- Grip O.
- Guilliams M.
- Malissen B.
- Agace W.W.
- Mowat A.M.
Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors.
However, monocytes are toll-like receptor responsive and during inflammation can differentiate into inflammatory macrophages.
19- Bain C.C.
- Scott C.L.
- Uronen-Hansson H.
- Gudjonsson S.
- Jansson O.
- Grip O.
- Guilliams M.
- Malissen B.
- Agace W.W.
- Mowat A.M.
Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors.
, 21- Grainger J.R.
- Wohlfert E.A.
- Fuss I.J.
- Bouladoux N.
- Askenase M.H.
- Legrand F.
- Koo L.Y.
- Brenchley J.M.
- Fraser I.D.
- Belkaid Y.
Inflammatory monocytes regulate pathologic responses to commensals during acute gastrointestinal infection.
In the dam-fed (DF) neonatal intestine, the embryonic macrophage–rich macrophage compartment predominates.
10- Bain C.C.
- Bravo-Blas A.
- Scott C.L.
- Gomez Perdiguero E.
- Geissmann F.
- Henri S.
- Malissen B.
- Osborne L.C.
- Artis D.
- Mowat A.M.
Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice.
Herein, we hypothesize that during NEC, monocytes are recruited and differentiated into monocyte-derived macrophages in the neonatal intestine in an NF-κB–dependent manner and this process contributes to NEC pathogenesis.
To test this hypothesis, IKKβ, the upstream kinase of NF-κB activation in myeloid cells, was deleted using lysozyme M (Lysm)Cre/+–IKKβf/f mice, and it was determined whether NEC development was affected in a neonatal mouse model. Using this strategy, IKKβ was targetedly deleted in monocytes, followed by examination of whether monocyte recruitment and differentiation into monocyte-derived macrophages was increased during NEC and affected by deletion of IKKβ.
Materials and Methods
Animal Experiments
LysmCre/+ IKKβf/f, Villin Cre+/− IKKβf/f, and IKKβf/f were kindly provided by Drs. Michael Karin and Lars Eckmann (University of California, San Diego, San Diego, CA).
B6.129(Cg)-Gt(ROSA)
26Sor
tm4(ACTB-tdTomato,-EGFP)Luo/J (mT/mG), Cx3cr1
GFP/+, and wild-type C57BL/6J (B6) mice were purchased from Jackson Laboratory (Bar Harbor, ME). NF-κB luciferase reporter mice were a gift from Fiona Yull (Vanderbilt University, Nashville, TN). NEC was initiated in mice within 24 hours of birth as follows: Pups were separated from the dams and placed into a 37°C humidified incubator (Air-Shield Vickers Medical, Hatboro, PA). At entry into the 72-hour NEC protocol, the mice were given a dose of 10
8 colony-forming unit bacteria from our standardized murine adult commensal bacterial preparation
22- Tian R.
- Liu S.X.
- Williams C.
- Soltau T.D.
- Dimmitt R.
- Zheng X.
- De Plaen I.G.
Characterization of a necrotizing enterocolitis model in newborn mice.
and 5 mg/kg lipopolysaccharide by gavage. Pups were gavage fed with Esbilac formula (PetAg, Inc., Hampshire, IL) every 3 hours and exposed to brief episodes of asphyxia (60 seconds in 100% N
2) followed by cold stress (4°C for 10 minutes) twice daily. Animals were observed for clinical signs of NEC, including severe abdominal distension, lethargy, and apnea. At signs of distress or at the end of the experimental period, animals were euthanized by decapitation. Tail biopsy was collected for genotyping. Survival time was recorded. Whole intestinal tissues were fixed in formalin, and tissue sections were stained with hematoxylin and eosin for evaluation by an investigator unaware of genotype results. Because the intestinal injury is typically patchy and scattered in this model, the entire intestine was fixed, sectioned, and examined. The injury was presented in two ways: both the grade and the extent of the injury. Because the prognosis of clinical NEC is determined by severe bowel injury, which leads to perforation and septic shock, the grade of histologic injury severity of the most affected area was first assigned after thorough examination of the bowel: grade I, injury limited to the tip of the villi; grade II, midvillous necrosis; grade III, complete villous necrosis; and grade IV, transmural necrosis. Severe NEC was defined as histologic grade ≥ II. The extent of the injury was then quantified. To calculate the injury score, the approximate percentage of each injured area was multiplied by its histologic injury index [1, tip of the villi, epithelial only; 2, villous injury with involvement of lamina propria (limited to mucosa); and 3, transmural necrosis]. The sum of the numbers obtained from each area was calculated for each pup. Pups that died before tissue collection were excluded from intestinal tissue assessment. Time points described refer to hours after NEC induction in experimental pups and their littermate DF controls. Specifically, 24-hour NEC and 24-hour DF controls are approximately 48 hours old and 48-hour NEC and 48-hour DF controls were approximately 72 hours old. For vascular perfusion studies, neonatal pups were anesthetized with 65 mg/kg of pentobarbital and injected by intracardiac infusion of 500 μL of 40 μg/mL Alexa Fluor–conjugated wheat germ agglutinin. Intestinal tissues were collected and fixed in formalin, paraffin embedded, and sectioned. Confocal vascular imaging was performed. The percentage of tissue section positive for wheat germ agglutinin was normalized to total tissue area using Fiji software version 1.51w (NIH, Bethesda, MD;
https://fiji.sc), and results were expressed as fold-changes to controls. To block L-selectin, 20 μg of a neutralizing antibody (clone MEL-14) or IgG control antibody (Biolegend, San Diego, CA) was injected intraperitoneally into wild-type pups 2 hours before NEC induction. mRNA relative fold-increases were determined compared with DF IgG-injected littermates, and percentage changes between IgG-injected NEC or L-selectin antibody–injected NEC were reported. Mice were maintained in the Stanley Manne Research Institute barrier facility, and experiments were approved by the Institutional Animal Care and Use Committee.
Immunohistochemistry/Immunofluorescence
Formalin-fixed, paraffin-embedded sections were stained using standard immunohistochemical methods for Cd31 (Abcam, Cambridge, MA) or green fluorescent protein (GFP; Abcam) Vina Green. Briefly, slides were deparaffinized in a series of xylene and alcohol washes, followed by antigen retrieval with heated citrate buffer. Endogenous peroxidase was blocked, and slides were incubated with primary antibody overnight at 4°C. Slides were washed and incubated for 1 hour with horseradish peroxidase–conjugated anti-rabbit or anti-chicken secondary antibody (Dako, Carpinteria, CA) to detect anti-Cd31 and anti-GFP antibodies, respectively. After washing, slides were developed using chromogen detection (Dako). Immunofluorescence for Selectin-L (SELL), CD11b, CX3CR1, and GFP was performed after citrate buffer retrieval using antibodies from Santa Cruz Biotechnology (Dallas, TX), Novus (Centennial, CO), Biolegend, and Abcam, respectively. Negative controls omitting the primary antibody and single-stained controls were performed concomitantly. Immunofluorescence terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining was performed using the In Situ Cell Death Detection Kit following the manufacturer's instructions (Sigma, St. Louis, MO), followed by incubation with an antibody against GFP. Slides were mounted with antifade gold with DAPI (Life Technologies, Grand Island, NY), and images were acquired using an LSM510 Zeiss confocal microscope (Oberkochen, Germany) or Leica DMR-HC upright microscope (Wetzlar, Germany). CD68 immunohistochemical staining of human sections was performed on unstained paraffin-embedded sections (4 μm thick). Slides were deparaffinized with serial xylene treatments and subjected to heat-induced epitope retrieval with cell conditioning solution 2 (Ventana Medical Systems, Tucson, AZ). Immunolabelling was performed using a mouse anti-CD68 monoclonal antibody (KP-1, prediluted) on the automated Ventana Benchmark XT system using the biotin-free Ventana Optiview DAB IHC Detection Kit (Ventana Medical Systems).
Real-Time RT-PCR
RNA was extracted using the RNeasy Mini Kit (Qiagen, Valencia, CA) following the manufacturer's instructions from tissues that were preserved in RNAlater (Qiagen) or from isolated cells. Reverse transcription was performed using the cDNA Archive kit (Life Technologies), and real-time PCR was performed using Power SYBR Green 2X master mix (Life Technologies) on an ABI 7500 Fast Real-Time PCR System (Thermo Fisher Scientifc, Waltham, MA). Primers spanned genomic DNA exon junctions to ensure specific amplification of mRNA. Sample values were normalized to glyceraldehyde-3-phosphate dehydrogenase, and fold-increases were calculated using the ΔΔCT method. Primer sequences were as follows: Ccl2, 5′-AAGCTGTAGTTTTTGTCACCAAGCT-3′ (forward) and 5′-TGGTTCCGATCCAGGTTTTTA-3′(reverse); IL-1β, 5′-TGACAGTGATGAGAATGACCTGTTC-3′ (forward) and 5′-GGACAGCCCAGGTCAAAGG-3′ (reverse); iNOS, 5′-CATCAGGTCGGCCATCACT-3′ (forward) and 5′-CGTACCGGATGAGCTGTGAA-3′ (reverse); TNF, 5′-GACCCTCACACTCAGATCATCTTCT-3′ (forward) and 5′-CCACTTGGTGGTTTGCTACGA-3′ (reverse); Il-6, 5′-TCGGAGGCTTAATTACACATGTTC-3′ (forward) and 5′-TGCCATTGCACAACTCTTTTCT-3′ (reverse); Mpo, 5′- CAGGACGTGAGGGTGACATG-3′ (forward) and 5′-GCTTCGTCTGTTGTTGCAGTGT-3′ (reverse); Mip2, 5′- TCAAGGGCGGTCAAAAAGTT-3′ (forward) and 5′-TTGCCTTTGTTCAGTATCTTTTGG-3′ (reverse); Mmp9, 5′-CCAGACGTGGGTCGATTCC-3′ (forward) and 5′-AGTAGTTTTGGATCCAGTATGTGATGTT-3′ (reverse); L-selectin, 5′-TGACGCCTGTCACAAACGA-3′ (forward) and 5′-GGCTGGCAAGAGGCTGTGT-3′ (reverse); and Itga4, 5′-CGACTTGAGAGGTGCTGTCTACATT-3′ (forward) and 5′-CAACGGCTACATCAACATATCCA-3′ (reverse).
LP Preparation and Flow Cytometry
To make lamina propria (LP) cell preparations, small intestines were cut longitudinally under a dissection microscope, carefully stripping away the mesenteric vasculature and pancreatic tissues. Tissue pieces were digested by shaking for 30 minutes at 37°C in phosphate-buffered saline supplemented with 5 mmol/L EDTA, 15 mmol/L HEPES, 1 mmol/L dithiothreitol, and 10% fetal bovine serum to release epithelial cells. Supernatants were discarded, and the tissues were washed in phosphate-buffered saline. The tissues were digested for 20 minutes in a Dulbecco's modified Eagle's medium buffer containing 1 mg/mL collagenase VIII (Sigma), 15 mmol/L HEPES, 0.1 mmol/L CaCl2, and 2% fetal bovine serum. Cells were filtered through a 40-μm strainer, centrifuged, washed, and enumerated. For flow cytometry, 1 × 106 cells were blocked in staining buffer (phosphate-buffered saline and 2% fetal bovine serum) with Fc block (Miltenyi Biotec, San Diego, CA) for 15 minutes on ice. Antibodies were then added for 20 minutes on ice. The antibody panel included Cd11b and MHC-II (BD Bioscience, Franklin Lakes, NJ); CD45 and F4/80 (ebioscience, San Diego, CA); and Ly6c, Ly6g, CD11c, and Cx3cr1 (Biolegend). Zombie live/dead discrimination (Biolegend) was used to gate out dead cells. When necessary for intracellular staining, cells were fixed and permeabilized (eBioscience), according to the manufacturer's instructions, and GFP was detected with anti-GFP antibody (Abcam). Data were collected on a BD LSR Fortessa and analyzed using FlowJo software version 10 (Tree Star, Ashland, OR). In the flow cytometry studies, Cd11b+ cells were defined as live, single, CD45+Cd11b+Ly6g−Side Scatterlo. For absolute cell counts CountBright absolute counting beads were added before flow cytometry acquisition, according to the manufacturer's instructions (Life Technologies).
Cell Isolation
Small intestinal LP cells were prepared, as described above, and incubated with biotin-labeled Cx3cr1 antibody (Biolegend), followed by anti-biotin microbeads, according to the manufacturer's instructions (Miltenyi Biotec). Cells were pelleted and lysed in RNA lysis buffer (Qiagen), and extraction was performed as described above.
Intestinal Permeability
Mouse litters were divided into two groups. One group was subjected to the NEC model for 24 hours, and the other group was left with the dam to be nursed (controls). At the end of the experiment, the pups in both groups were fasted for 2 hours, and then received 750 mg/kg of 10-kDa fluorescein isothiocyanate–dextran (FD-10S; Sigma) via gavage. Four hours later, blood samples were collected for fluorescence determination and the tails of the mice were biopsied for genotyping. Serum fluorescence was measured with a fluorescence spectrophotometer, and fluorescein isothiocyanate–dextran serum concentration was determined by comparison to a standard curve of known fluorescein isothiocyanate–dextran concentrations.
Luciferase Assay
Tails and small intestinal tissues were homogenized in 1 mL of lysis buffer. Freshly reconstituted luciferase assay buffer (100 μL) was added to 20 μL of tissue homogenate, and luciferase activity was detected with a luciferase assay system (Promega, Madison, WI). Results were expressed as relative light units normalized to intestinal protein concentration, as measured by the Bradford assay.
In Vivo Imaging
Pups were given luciferin (Gold Biotechnology, St. Louis, MO), 0.1 mg each, intraperitoneally, 10 minutes before each imaging. Pups were gently restrained with tape while bioluminescence images were taken using an intensified charge-coupled device camera (Xenogen IVIS imaging system; Xenogen Corp., Hopkinton, MA). Images were obtained at low binning for 5 seconds at the F8-sensitivity threshold, as these parameters were found to best detect intensity differences in neonatal pups while avoiding color saturation during image acquisition. A standard area over the abdomen was defined and used to analyze integrated photon intensity.
Statistical Analysis
Two-tailed t-test was used to evaluate differences between the two groups. When more than two groups were compared, analysis of variance analysis with a Tukey post test was performed. To evaluate differences in the incidence of severe NEC (grade ≥II), χ2 analysis was used. Animal survival data were analyzed by log-rank test. Differences were considered statistically significant when P ≤ 0.05. Error was calculated using SEM.
Discussion
Herein, it was demonstrated that in the murine intestine during NEC, monocytes undergo differentiation into macrophages via an IKKβ-dependent mechanism. This differentiation process is associated with up-regulation of monocyte-associated gene expression in the intestinal macrophage compartment. It was prevented when monocyte adhesion to endothelial cells was inhibited by L-selectin antibody. Pups with Lysm-monocyte IKKβ deletion had improved survival and decreased incidence of severe NEC in comparison to their littermate controls. In human NEC specimens, a higher number of myeloid cells and SELL+ cells were found compared with control intestinal tissues. Together, these findings support a role for recruitment of Ly6c+ monocytes to the intestine and Ly6c+ monocyte differentiation into macrophages in NEC development, in a process mediated by NF-κB signaling.
This study, along with several previous studies, has observed that the cellular inflammatory response resulted in macrophage-rich infiltrates of intestinal tissues in human NEC
27- Remon J.I.
- Amin S.C.
- Mehendale S.R.
- Rao R.
- Luciano A.A.
- Garzon S.A.
- Maheshwari A.
Depth of bacterial invasion in resected intestinal tissue predicts mortality in surgical necrotizing enterocolitis.
, 28- Pender S.L.
- Braegger C.
- Gunther U.
- Monteleone G.
- Meuli M.
- Schuppan D.
- Macdonald T.T.
Matrix metalloproteinases in necrotising enterocolitis.
, 29- Ballance W.A.
- Dahms B.B.
- Shenker N.
- Kliegman R.M.
Pathology of neonatal necrotizing enterocolitis: a ten-year experience.
and also in neonatal mouse NEC models. Most of this previous research dealt with late stages of disease when intestinal injury was already present.
3- MohanKumar K.
- Kaza N.
- Jagadeeswaran R.
- Garzon S.A.
- Bansal A.
- Kurundkar A.R.
- Namachivayam K.
- Remon J.I.
- Bandepalli C.R.
- Feng X.
- Weitkamp J.H.
- Maheshwari A.
Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model.
, 30- Ginzel M.
- Feng X.
- Kuebler J.F.
- Klemann C.
- Yu Y.
- von Wasielewski R.
- Park J.K.
- Hornef M.W.
- Vieten G.
- Ure B.M.
- Kaussen T.
- Gosemann J.H.
- Mayer S.
- Suttkus A.
- Lacher M.
Dextran sodium sulfate (DSS) induces necrotizing enterocolitis-like lesions in neonatal mice.
Herein, the initial 24 hours during NEC development were studied to identify the formative changes in the myeloid compartment before disease. This early time point not only helps to discriminate the recently arrived monocytes but also allows the interrogation of NF-κB signaling events that may occur before robust up-regulation of inflammatory genes at 48 hours when myeloid cells are increased. NF-κB–mediated recruitment and differentiation of monocytes occurred within 24 hours into the NEC model. This process was mediated via L-selectin, suggesting that monocyte adhesion was one of the pathways involved. The significance of these data is strengthened by the recent study that identified nongranulocyte adhesion and diapedesis as top canonical pathways involved in human NEC.
31- MohanKumar K.
- Namachivayam K.
- Cheng F.
- Jiang R.H.
- Flores-Torres J.
- Torres B.A.
- Maheshwari A.
Trinitrobenzene sulfonic acid-induced intestinal injury in neonatal mice activates transcriptional networks similar to those seen in human necrotizing enterocolitis.
Several studies have demonstrated a specific role for monocytes in promoting NEC. Mechanisms reported include the promotion of type 17 helper T-cell differentiation that leads to an imbalance between type 17 helper T cells and regulatory T cells
5- Pang Y.
- Du X.
- Xu X.
- Wang M.
- Li Z.
Monocyte activation and inflammation can exacerbate Treg/Th17 imbalance in infants with neonatal necrotizing enterocolitis.
and increased M1 macrophage polarization that leads to an increase in IEC apoptosis.
6M1 to M2 macrophage polarization in heparin-binding epidermal growth factor-like growth factor therapy for necrotizing enterocolitis.
Myeloid cells may also be protective in NEC. A recent study showed that immunosuppressive myeloid-derived suppressor cells are found in the spleen and bone marrow of neonatal mice and humans, which potently inhibit proliferation of CD4
+ and CD8
+ T cells. Myeloid-derived suppressor cells were identified at days 4 through 14 of life and play a protective role in controlling experimental NEC in 4-day–old mice.
4- He Y.M.
- Li X.
- Perego M.
- Nefedova Y.
- Kossenkov A.V.
- Jensen E.A.
- Kagan V.
- Liu Y.F.
- Fu S.Y.
- Ye Q.J.
- Zhou Y.H.
- Wei L.
- Gabrilovich D.I.
- Zhou J.
Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation.
Intriguingly, these cells were not present in the first 3 days, which the authors suggest is a period of increased NEC susceptibility.
4- He Y.M.
- Li X.
- Perego M.
- Nefedova Y.
- Kossenkov A.V.
- Jensen E.A.
- Kagan V.
- Liu Y.F.
- Fu S.Y.
- Ye Q.J.
- Zhou Y.H.
- Wei L.
- Gabrilovich D.I.
- Zhou J.
Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation.
Whether macrophages derived from monocytes, which were found to be recruited during NEC in an IKKβ-dependent manner, impact the T-cell compartment remains unknown.
Our laboratory has reported that an increase in epithelial barrier permeability precedes NEC in this mouse model.
2- Bergmann K.R.
- Liu S.X.
- Tian R.
- Kushnir A.
- Turner J.R.
- Li H.L.
- Chou P.M.
- Weber C.R.
- De Plaen I.G.
Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis.
Increased intestinal permeability has been associated with other NEC models
32- Hackam D.J.
- Upperman J.S.
- Grishin A.
- Ford H.R.
Disordered enterocyte signaling and intestinal barrier dysfunction in the pathogenesis of necrotizing enterocolitis.
, 33- White J.R.
- Gong H.
- Pope B.
- Schlievert P.
- McElroy S.J.
Paneth-cell-disruption-induced necrotizing enterocolitis in mice requires live bacteria and occurs independently of TLR4 signaling.
and with human NEC.
34- Moore S.A.
- Nighot P.
- Reyes C.
- Rawat M.
- McKee J.
- Lemon D.
- Hanson J.
- Ma T.Y.
Intestinal barrier dysfunction in human necrotizing enterocolitis.
Herein, it was shown that increased intestinal permeability and decreased epithelial proliferation in NEC were present independently of IKKβ status in Lysm monocytes. Because intestinal injury was markedly attenuated in Lysm-IKKβ–deleted NEC mice, increased intestinal permeability may be necessary but not sufficient to induce intestinal injury in this NEC model. In addition, these findings suggest that therapies targeting monocyte NF-κB signaling independently of epithelial effects may be promising in NEC.
Recent evidence challenges the current paradigm involving the adult small intestinal macrophage pool. Data suggest that this compartment is composed of three distinct subsets of equal proportions.
35- Shaw T.N.
- Houston S.A.
- Wemyss K.
- Bridgeman H.M.
- Barbera T.A.
- Zangerle-Murray T.
- Strangward P.
- Ridley A.J.L.
- Wang P.
- Tamoutounour S.
- Allen J.E.
- Konkel J.E.
- Grainger J.R.
Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression.
Two of these populations are derived from blood monocytes but have different replenishment rates and are phenotypically described as Tim4
−CD4
+ (slow rate) and Tim4
−CD4
− (high rate). These populations both transition from Ly6c
+ monocytes to Ly6c
− macrophages through differentiation mechanisms described as the monocyte waterfall,
19- Bain C.C.
- Scott C.L.
- Uronen-Hansson H.
- Gudjonsson S.
- Jansson O.
- Grip O.
- Guilliams M.
- Malissen B.
- Agace W.W.
- Mowat A.M.
Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors.
, 36- Tamoutounour S.
- Henri S.
- Lelouard H.
- de Bovis B.
- de Haar C.
- van der Woude C.J.
- Woltman A.M.
- Reyal Y.
- Bonnet D.
- Sichien D.
- Bain C.C.
- Mowat A.M.
- Reis e Sousa C.
- Poulin L.F.
- Malissen B.
- Guilliams M.
CD64 distinguishes macrophages from dendritic cells in the gut and reveals the Th1-inducing role of mesenteric lymph node macrophages during colitis.
, 37- Zigmond E.
- Varol C.
- Farache J.
- Elmaliah E.
- Satpathy A.T.
- Friedlander G.
- Mack M.
- Shpigel N.
- Boneca I.G.
- Murphy K.M.
- Shakhar G.
- Halpern Z.
- Jung S.
Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells.
which is named for the appearance of the flow cytometry plot as cells lose Ly6c and gain MHC-II or Cx3cr1. This waterfall is typically absent in the neonatal intestine, but it is apparent in the adult, where the continuous replacement of most intestinal macrophages by bone marrow–derived monocytes helps achieve the precise balance between tolerance and immune activation. The third population, previously thought to be replaced before reaching adulthood,
10- Bain C.C.
- Bravo-Blas A.
- Scott C.L.
- Gomez Perdiguero E.
- Geissmann F.
- Henri S.
- Malissen B.
- Osborne L.C.
- Artis D.
- Mowat A.M.
Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice.
is the embryonically derived Tim-4
+CD4
+ population, and it is locally maintained independently of monocytes (embryonic macrophages).
35- Shaw T.N.
- Houston S.A.
- Wemyss K.
- Bridgeman H.M.
- Barbera T.A.
- Zangerle-Murray T.
- Strangward P.
- Ridley A.J.L.
- Wang P.
- Tamoutounour S.
- Allen J.E.
- Konkel J.E.
- Grainger J.R.
Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression.
In the intestine during the neonatal period, the Tim-4
+CD4
+ (embryonic macrophage) predominates
35- Shaw T.N.
- Houston S.A.
- Wemyss K.
- Bridgeman H.M.
- Barbera T.A.
- Zangerle-Murray T.
- Strangward P.
- Ridley A.J.L.
- Wang P.
- Tamoutounour S.
- Allen J.E.
- Konkel J.E.
- Grainger J.R.
Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression.
and Ly6c
+ monocytes are rare.
10- Bain C.C.
- Bravo-Blas A.
- Scott C.L.
- Gomez Perdiguero E.
- Geissmann F.
- Henri S.
- Malissen B.
- Osborne L.C.
- Artis D.
- Mowat A.M.
Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice.
These data concur with our findings describing a low number of Ly6c
+ cells/Lysm
+ cells in the neonatal intestine.
The gut embryonic macrophage population maintains the integrity of enteric neurons as well as submucosal and mucosal microvasculature in adults.
38- De Schepper S.
- Verheijden S.
- Aguilera-Lizarraga J.
- Viola M.F.
- Boesmans W.
- Stakenborg N.
- Voytyuk I.
- Smidt I.
- Boeckx B.
- Dierckx de Casterle I.
- Baekelandt V.
- Gonzalez Dominguez E.
- Mack M.
- Depoortere I.
- De Strooper B.
- Sprangers B.
- Himmelreich U.
- Soenen S.
- Guilliams M.
- Vanden Berghe P.
- Jones E.
- Lambrechts D.
- Boeckxstaens G.
Self-maintaining gut macrophages are essential for intestinal homeostasis.
Depletion of the embryonic macrophage population via diphtheria toxin in adult transgenic mice resulted in their replacement by blood monocytes that differentiated into gut Cx3cr1
+ monocyte-derived macrophages
38- De Schepper S.
- Verheijden S.
- Aguilera-Lizarraga J.
- Viola M.F.
- Boesmans W.
- Stakenborg N.
- Voytyuk I.
- Smidt I.
- Boeckx B.
- Dierckx de Casterle I.
- Baekelandt V.
- Gonzalez Dominguez E.
- Mack M.
- Depoortere I.
- De Strooper B.
- Sprangers B.
- Himmelreich U.
- Soenen S.
- Guilliams M.
- Vanden Berghe P.
- Jones E.
- Lambrechts D.
- Boeckxstaens G.
Self-maintaining gut macrophages are essential for intestinal homeostasis.
and led to reduced abundance of the intestinal microvasculature and decreased enteric neuron density.
38- De Schepper S.
- Verheijden S.
- Aguilera-Lizarraga J.
- Viola M.F.
- Boesmans W.
- Stakenborg N.
- Voytyuk I.
- Smidt I.
- Boeckx B.
- Dierckx de Casterle I.
- Baekelandt V.
- Gonzalez Dominguez E.
- Mack M.
- Depoortere I.
- De Strooper B.
- Sprangers B.
- Himmelreich U.
- Soenen S.
- Guilliams M.
- Vanden Berghe P.
- Jones E.
- Lambrechts D.
- Boeckxstaens G.
Self-maintaining gut macrophages are essential for intestinal homeostasis.
During development, embryonic macrophages play a role in vascular maturation and remodeling
14- Leid J.
- Carrelha J.
- Boukarabila H.
- Epelman S.
- Jacobsen S.E.
- Lavine K.J.
Primitive embryonic macrophages are required for coronary development and maturation.
, 15- De Kleer I.
- Willems F.
- Lambrecht B.
- Goriely S.
Ontogeny of myeloid cells.
, 16- DeFalco T.
- Bhattacharya I.
- Williams A.V.
- Sams D.M.
- Capel B.
Yolk-sac-derived macrophages regulate fetal testis vascularization and morphogenesis.
, 17- Fantin A.
- Vieira J.M.
- Gestri G.
- Denti L.
- Schwarz Q.
- Prykhozhij S.
- Peri F.
- Wilson S.W.
- Ruhrberg C.
Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction.
and in the removal of debris (such as dying epithelial cells).
11- Stevens B.
- Allen N.J.
- Vazquez L.E.
- Howell G.R.
- Christopherson K.S.
- Nouri N.
- Micheva K.D.
- Mehalow A.K.
- Huberman A.D.
- Stafford B.
- Sher A.
- Litke A.M.
- Lambris J.D.
- Smith S.J.
- John S.W.
- Barres B.A.
The classical complement cascade mediates CNS synapse elimination.
, 12Macrophages are required for cell death and tissue remodeling in the developing mouse eye.
, 13- Varol C.
- Mildner A.
- Jung S.
Macrophages: development and tissue specialization.
The intestinal microvasculature develops tremendously during the neonatal period,
39- Stappenbeck T.S.
- Hooper L.V.
- Gordon J.I.
Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells.
and NEC stresses decrease intestinal microvasculature density.
40- Yan X.
- Managlia E.
- Liu S.X.
- Tan X.D.
- Wang X.
- Marek C.
- De Plaen I.G.
Lack of VEGFR2 signaling causes maldevelopment of the intestinal microvasculature and facilitates necrotizing enterocolitis in neonatal mice.
A premature influx of monocyte-derived macrophages in the neonatal intestine induced by NEC stresses may disturb the embryonic macrophage compartment at a time of intense development of the intestinal microvasculature and likely impact the environment necessary to orchestrate its maturation at this stage. In support of this idea, it was demonstrated that the intestinal microvascular density was preserved in Lysm-IKKβ–deficient mice during NEC versus their IKKβ-sufficient littermates (
Supplemental Figure S4, E and F). Further studies are needed to determine whether the early influx of monocyte-derived macrophages into the intestine impacts the self-maintaining gut macrophage compartment, thus affecting microvasculature development.
During inflammation, recruited monocytes adhere to activated endothelial cells. This interaction induces MHC-II expression, leading to monocyte differentiation and subsequent tissue infiltration.
18- Jakubzick C.
- Gautier E.L.
- Gibbings S.L.
- Sojka D.K.
- Schlitzer A.
- Johnson T.E.
- Ivanov S.
- Duan Q.
- Bala S.
- Condon T.
- van Rooijen N.
- Grainger J.R.
- Belkaid Y.
- Ma'ayan A.
- Riches D.W.
- Yokoyama W.M.
- Ginhoux F.
- Henson P.M.
- Randolph G.J.
Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes.
This process sequentially involves down-regulation of Ly6c and Ccr2 and up-regulation of Cx3cr1.
20Macrophages in intestinal homeostasis and inflammation.
Herein, it was shown that genes preferentially associated with monocytes but not embryonic macrophages were up-regulated during NEC in the Cx3cr1
+ population in IKKβ-sufficient but not in Lysm-IKKβ–deleted pups. When monocyte adhesion was blocked by L-selectin antibody, the Cx3cr1 population had decreased expression of these genes. Because Cx3cr1 is expressed at high level in yolk sac macrophages
41- Yona S.
- Kim K.W.
- Wolf Y.
- Mildner A.
- Varol D.
- Breker M.
- Strauss-Ayali D.
- Viukov S.
- Guilliams M.
- Misharin A.
- Hume D.A.
- Perlman H.
- Malissen B.
- Zelzer E.
- Jung S.
Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis.
but not in fetal liver monocytes before tissue infiltration,
25- Hoeffel G.
- Chen J.
- Lavin Y.
- Low D.
- Almeida F.F.
- See P.
- Beaudin A.E.
- Lum J.
- Low I.
- Forsberg E.C.
- Poidinger M.
- Zolezzi F.
- Larbi A.
- Ng L.G.
- Chan J.K.
- Greter M.
- Becher B.
- Samokhvalov I.M.
- Merad M.
- Ginhoux F.
C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages.
our findings demonstrate that the intestinal macrophage compartment was enriched in monocyte-derived macrophages during NEC in IKKβ-sufficient pups but not in Lysm-IKKβ–deleted pups and that, in the intestine, NF-κB activation up-regulated the target genes
L-selectin and
Mmp9 in monocytes. More important, the number of SELL
+ cells was also increased in the villi of human NEC tissue specimens compared with controls, which further supports monocyte recruitment in NEC pathogenesis.
Several studies have investigated the gene signatures of both macrophages derived from monocytes and embryonic macrophages
14- Leid J.
- Carrelha J.
- Boukarabila H.
- Epelman S.
- Jacobsen S.E.
- Lavine K.J.
Primitive embryonic macrophages are required for coronary development and maturation.
, 25- Hoeffel G.
- Chen J.
- Lavin Y.
- Low D.
- Almeida F.F.
- See P.
- Beaudin A.E.
- Lum J.
- Low I.
- Forsberg E.C.
- Poidinger M.
- Zolezzi F.
- Larbi A.
- Ng L.G.
- Chan J.K.
- Greter M.
- Becher B.
- Samokhvalov I.M.
- Merad M.
- Ginhoux F.
C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages.
, 26- Pucci F.
- Venneri M.A.
- Biziato D.
- Nonis A.
- Moi D.
- Sica A.
- Di Serio C.
- Naldini L.
- De Palma M.
A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophages suggests common functions and developmental relationships.
and found
Ly6c,
Ccl2,
Lysm,
Mmp9, and
L-selectin to be strongly associated with monocyte-derived cells, whereas
Cx3cr1 was associated with an embryonic signature. This aligns with the finding that Lysm was expressed in Ly6c
+ cells. However, it was surprising to find a lack of Lysm expression in most neonatal intestinal myeloid cells, especially given that Lysm-expressing cells are abundantly present in both their liver and spleen (
Supplemental Figure S4D) as well as in the adult intestine (
Supplemental Figure S4C). These data demonstrate that in the neonatal period, unlike the adult,
23- Clausen B.E.
- Burkhardt C.
- Reith W.
- Renkawitz R.
- Forster I.
Conditional gene targeting in macrophages and granulocytes using LysMcre mice.
, 24- Cross M.
- Mangelsdorf I.
- Wedel A.
- Renkawitz R.
Mouse lysozyme M gene: isolation, characterization, and expression studies.
Lysm recombination should not been used to target the global intestinal myeloid population because its expression is mainly in monocyte-derived cells. Furthermore, it shows that in steady state, these Lysm
+ cells are normally excluded from the intestine.
Together, our findings support a role for monocyte adhesion and recruitment to intestinal tissue during NEC in a process mediated by NF-κB signaling at a time when typically the intestinal tissue myeloid compartment is mostly constituted by embryonic macrophages. Preventing early monocyte recruitment to the neonatal intestine may be a novel and an effective strategy to prevent intestinal injury in NEC.
Article info
Publication history
Published online: December 26, 2018
Accepted:
November 28,
2018
Footnotes
Supported by NIH grants R01GM122406 and R01GM117628 (X.-D.T.), 2RO1 DK095662 (T.A.B.), and R01HD060876 (I.G.D.P.); Veterans Affairs Merit grant 1I01CX001353-01 (T.A.B.); Northwestern University Flow Cytometry Facility and National Cancer Institute grant CA060553; and the Stanley Manne Children's Research Institute of the Ann & Robert H. Lurie Children's Hospital of Chicago (I.G.D.P.).
Disclosures: None declared.
Copyright
© 2019 American Society for Investigative Pathology. Published by Elsevier Inc.