51
|
Burge K, Bergner E, Gunasekaran A, Eckert J, Chaaban H. The Role of Glycosaminoglycans in Protection from Neonatal Necrotizing Enterocolitis: A Narrative Review. Nutrients 2020; 12:nu12020546. [PMID: 32093194 PMCID: PMC7071410 DOI: 10.3390/nu12020546] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/22/2022] Open
Abstract
Necrotizing enterocolitis, a potentially fatal intestinal inflammatory disorder affecting primarily premature infants, is a significant cause of morbidity and mortality in neonates. While the etiology of the disease is, as yet, unknown, a number of risk factors for the development of necrotizing enterocolitis have been identified. One such risk factor, formula feeding, has been shown to contribute to both increased incidence and severity of the disease. The protective influences afforded by breastfeeding are likely attributable to the unique composition of human milk, an extremely potent, biologically active fluid. This review brings together knowledge on the pathogenesis of necrotizing enterocolitis and current thinking on the instrumental role of one of the more prominent classes of bioactive components in human breast milk, glycosaminoglycans.
Collapse
MESH Headings
- Breast Feeding
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/prevention & control
- Female
- Glycosaminoglycans/pharmacology
- Humans
- Infant Formula/adverse effects
- Infant, Newborn
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/pathology
- Infant, Premature, Diseases/prevention & control
- Male
- Milk, Human/chemistry
- Protective Agents/pharmacology
- Risk Factors
Collapse
|
52
|
Elevated Coefficient of Variation in Total Fecal Bile Acids Precedes Diagnosis of Necrotizing Enterocolitis. Sci Rep 2020; 10:249. [PMID: 31937876 PMCID: PMC6959237 DOI: 10.1038/s41598-019-57178-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022] Open
Abstract
Accumulation of bile acids (BAs) may mediate development of necrotizing enterocolitis (NEC). Serial fecal samples were collected from premature infants with birth weight (BW) ≤ 1800 g, estimated gestational age (EGA) ≤ 32 weeks, and <30 days old prior to initiation of enteral feeding. Nine infants that developed Bell’s Stage ≥ II NEC were matched with control infants based on BW, EGA, day of life (DOL) enteral feeding was initiated and DOL of the first sample. From each subject, five samples matched by DOL collected were analyzed for BA levels and composition. Fifteen individual BA species were measured via LC-MS/MS and total BA levels were measured using the Diazyme Total Bile Acid Assay kit. No statistically significant differences in composition were observed between control and NEC at the level of individual species (p = 0.1133) or grouped BAs (p = 0.0742). However, there was a statistically significant difference (p = 0.000012) in the mean coefficient of variation (CV) between the two groups with infants developing NEC having more than four-fold higher mean CV than controls. Importantly, these variations occurred prior to NEC diagnosis. These data suggest fluctuations in total fecal BA levels could provide the basis for the first predictive clinical test for NEC.
Collapse
|
53
|
Alinaghi M, Nguyen DN, Sangild PT, Bertram HC. Direct Implementation of Intestinal Permeability Test in NMR Metabolomics for Simultaneous Biomarker Discovery-A Feasibility Study in a Preterm Piglet Model. Metabolites 2020; 10:metabo10010022. [PMID: 31906404 PMCID: PMC7022985 DOI: 10.3390/metabo10010022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/22/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022] Open
Abstract
Measurement of intestinal permeability (IP) is often used in the examination of inflammatory gastrointestinal disorders. IP can be assessed by measurement of urinary recovery of ingested non-metabolizable lactulose (L) and mannitol (M). The present study aimed to examine how measurements of IP can be integrated in a NMR-based metabolomics approach for a simultaneous quantification of L/M ratio and biomarker exploration. For this purpose, plasma and urine samples were collected from five-day-old preterm piglets (n = 20) with gastrointestinal disorders (subjected to intra-amniotic lipopolysaccharide (LPS, 1 mg/fetus)) after they had been administrated a 5% lactulose and 5% mannitol solution (15 mL/kg). The collected plasma and urine samples were analyzed by 1H NMR-based metabolomics. Urine L/M ratio measured by 1H NMR spectroscopy showed high correlation with the standard measurement of the urinary recoveries by enzymatic assays (r = 0.93, p < 0.05). Partial least squares (PLS) regressions and correlation analyses between L/M ratio and NMR metabolomics data revealed that L/M ratio was positively correlated with plasma lactate, acetate and succinate levels and negatively correlated with urinary hippuric acid and glycine. In conclusion, the present study demonstrated that NMR metabolomics enables simultaneous IP testing and discovery of biomarkers associated with an impaired intestinal permeability.
Collapse
Affiliation(s)
- Masoumeh Alinaghi
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev, Denmark;
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlaegevej 68, 1860 Frederiksberg C, Denmark; (D.N.N.); (P.T.S.)
| | - Per Torp Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlaegevej 68, 1860 Frederiksberg C, Denmark; (D.N.N.); (P.T.S.)
| | - Hanne Christine Bertram
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev, Denmark;
- Correspondence:
| |
Collapse
|
54
|
Palumbo P, Lombardi F, Cifone MG, Cinque B. The Epithelial Barrier Model Shows That the Properties of VSL#3 Depend from Where it is Manufactured. Endocr Metab Immune Disord Drug Targets 2019; 19:199-206. [PMID: 30360752 PMCID: PMC6425067 DOI: 10.2174/1871530318666181022164505] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/29/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
Abstract
Background: VSL#3 has been extensively investigated and is currently recommended for the prevention and treatment of chronic pouchitis and ulcerative colitis. Nonetheless, in vitro and in vivo stud-ies have recently shown variability in the VSL#3 efficacy often attributed to the manufacturing process. Objective: The aim was to comparatively study the in vitro effects of two VSL#3 preparations produced in different sites (named US- and Italy-made VSL#3) on CaCo-2 epithelial barrier model in terms of trans-epithelial electrical resistance (TEER), dextran flux and expression of Tight Junctions (TJ) proteins i.e. zonulin-1 (ZO-1) and occludin, in the absence or presence of a heat stress-related damage of mono-layer. Methods: TEER was evaluated on CaCo-2 differentiated monolayers. Epithelial permeability of polarized monolayers was assessed by measuring the FITC-labeled dextran flux from the apical to basolateral chambers. ZO-1/occludin levels were analyzed by western blot analysis. A set of experiments was per-formed to compare the effects of both VSL#3 on TEER values, dextran flux and ZO-1/occludin expres-sion in CaCo-2 monolayers after heat stress exposure. Results: US- and Italy-made VSL#3 have opposing effects on TEER values, dextran flux, and ZO-1/occludin expression, being all these parameters negatively influenced just by Italy-made product. US-made probiotic did not affect baseline TEER, dextran flux and ZO-1 expression and strongly increased occludin levels. Of note, pre-treatment of monolayer with US-made VSL#3, but not Italy-made product, totally prevented the heat-induced epithelial barrier integrity loss. Conclusion: Our data trigger the need for reassessing efficacy or safety of the Italy-made VSL#3 con-sidering intestinal epithelial barrier plays an important role in maintaining host health.
Collapse
Affiliation(s)
- Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila - Building Delta 6, Coppito, L'Aquila, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila - Building Delta 6, Coppito, L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila - Building Delta 6, Coppito, L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila - Building Delta 6, Coppito, L'Aquila, Italy
| |
Collapse
|
55
|
Ares GJ, Buonpane C, Yuan C, Wood D, Hunter CJ. A Novel Human Epithelial Enteroid Model of Necrotizing Enterocolitis. J Vis Exp 2019. [PMID: 31033943 DOI: 10.3791/59194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease of newborn infants. It is characterized by multiple pathophysiologic alterations in the human intestinal epithelium, leading to increased intestinal permeability, impaired restitution, and increased cell death. Although there are numerous animal models of NEC, response to injury and therapeutic interventions may be highly variable between species. Furthermore, it is ethically challenging to study disease pathophysiology or novel therapeutic agents directly in human subjects, especially children. Therefore, it is highly desirable to develop a novel model of NEC using human tissue. Enteroids are 3-dimensional organoids derived from intestinal epithelial cells. They are ideal for the study of complex physiologic interactions, cell signaling, and host-pathogen defense. In this manuscript we describe a protocol that cultures human enteroids after isolating intestinal stem cells from patients undergoing bowel resection. The crypt cells are cultured in media containing growth factors that encourage differentiation into the various cell types native of the human intestinal epithelium. These cells are grown in a synthetic, collagenous mix of proteins that serve as a scaffold, mimicking the extra-cellular basement membrane. As a result, enteroids develop apical-basolateral polarity. Co-administration of lipopolysaccharide (LPS) in media causes an inflammatory response in the enteroids, leading to histologic, genetic, and protein expression alterations similar to those seen in human NEC. An experimental model of NEC using human tissue may provide a more accurate platform for drug and treatment testing prior to human trials, as we strive to identify a cure for this disease.
Collapse
Affiliation(s)
| | | | - Carrie Yuan
- Department of Pediatrics, Northwestern University
| | - Douglas Wood
- Department of Pediatrics, Northwestern University
| | - Catherine J Hunter
- Department of Pediatrics, Northwestern University; Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago;
| |
Collapse
|
56
|
Ares G, Buonpane C, Sincavage J, Yuan C, Wood DR, Hunter CJ. Caveolin 1 is Associated with Upregulated Claudin 2 in Necrotizing Enterocolitis. Sci Rep 2019; 9:4982. [PMID: 30899070 PMCID: PMC6428816 DOI: 10.1038/s41598-019-41442-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency of neonates. Epithelial tight junction (TJ) proteins, such as claudins, are essential for regulation and function of the intestinal barrier. Rho kinase (ROCK) affects cellular permeability and TJ regulation. We hypothesized that TJ protein changes would correlate with increased permeability in experimental NEC, and ROCK inhibitors would be protective against NEC by regulation of key claudin proteins. We tested this hypothesis using an in vivo rat pup model, an in vitro model of experimental NEC, and human intestinal samples from patients with and without NEC. Experimental NEC was induced in rats via hypoxia and bacteria-containing formula, and in Caco-2 cells by media inoculated with LPS. The expression of claudins was measured by gene and protein analysis. Experimental NEC in rat pups and Caco-2 cells had increased permeability compared to controls. Gene and protein expression of claudin 2 was increased in experimental NEC. Sub-cellular fractionation localized increased claudin 2 protein to the cytoskeleton. ROCK inhibition was associated with normalization of these alterations and decreased severity of experimental NEC. Co-immunoprecipitation of caveolin-1 with claudin 2 suggests that caveolin-1 may act as a shuttle for the internalization of claudin 2 seen in experimental NEC. In conclusion, NEC is associated with intestinal permeability and increased expression of claudin 2, increased binding of caveolin-1 and claudin 2, and increased trafficking of claudin 2 to the cytoskeleton.
Collapse
Affiliation(s)
- Guillermo Ares
- University of Illinois at Chicago, Department of Surgery, 840S Wood Street, Suite 376-CSN, Chicago, IL, 60612, USA.,Feinberg School of Medicine, Northwestern University, Department of Pediatrics, 310 East Superior - Morton 4-685, Chicago, IL, 60611, USA
| | - Christie Buonpane
- Feinberg School of Medicine, Northwestern University, Department of Pediatrics, 310 East Superior - Morton 4-685, Chicago, IL, 60611, USA.,Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Avenue, Box 63, Chicago, IL, 60611, USA
| | - John Sincavage
- Feinberg School of Medicine, Northwestern University, Department of Pediatrics, 310 East Superior - Morton 4-685, Chicago, IL, 60611, USA
| | - Carrie Yuan
- Feinberg School of Medicine, Northwestern University, Department of Pediatrics, 310 East Superior - Morton 4-685, Chicago, IL, 60611, USA
| | - Douglas R Wood
- Feinberg School of Medicine, Northwestern University, Department of Pediatrics, 310 East Superior - Morton 4-685, Chicago, IL, 60611, USA
| | - Catherine J Hunter
- Feinberg School of Medicine, Northwestern University, Department of Pediatrics, 310 East Superior - Morton 4-685, Chicago, IL, 60611, USA. .,Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Avenue, Box 63, Chicago, IL, 60611, USA.
| |
Collapse
|
57
|
Arthur CM, Nalbant D, Feldman HA, Saeedi BJ, Matthews J, Robinson BS, Kamili NA, Bennett A, Cress GA, Sola-Visner M, Jones RM, Zimmerman MB, Neish AS, Patel RM, Nopoulos P, Georgieff MK, Roback JD, Widness JA, Josephson CD, Stowell SR. Anemia induces gut inflammation and injury in an animal model of preterm infants. Transfusion 2019; 59:1233-1245. [PMID: 30897226 DOI: 10.1111/trf.15254] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND While very low birth weight (VLBW) infants often require multiple red blood cell transfusions, efforts to minimize transfusion-associated risks have resulted in more restrictive neonatal transfusion practices. However, whether restrictive transfusion strategies limit transfusions without increasing morbidity and mortality in this population remains unclear. Recent epidemiologic studies suggest that severe anemia may be an important risk factor for the development of necrotizing enterocolitis (NEC). However, the mechanism whereby anemia may lead to NEC remains unknown. STUDY DESIGN AND METHODS The potential impact of anemia on neonatal inflammation and intestinal barrier disruption, two well-characterized predisposing features of NEC, was defined by correlation of hemoglobin values to cytokine levels in premature infants and by direct evaluation of intestinal hypoxia, inflammation and gut barrier disruption using a pre-clinical neonatal murine model of phlebotomy-induced anemia (PIA). RESULTS Increasing severity of anemia in the preterm infant correlated with the level of IFN-gamma, a key pro-inflammatory cytokine that may predispose an infant to NEC. Gradual induction of PIA in a pre-clinical model resulted in significant hypoxia throughout the intestinal mucosa, including areas where intestinal macrophages reside. PIA-induced hypoxia significantly increased macrophage pro-inflammatory cytokine levels, while reducing tight junction protein ZO-1 expression and increasing intestinal barrier permeability. Macrophage depletion reversed the impact of anemia on intestinal ZO-1 expression and barrier function. CONCLUSIONS Taken together, these results suggest that anemia can increase intestinal inflammation and barrier disruption likely through altered macrophage function, leading to the type of predisposing intestinal injury that may increase the risk for NEC.
Collapse
Affiliation(s)
- Connie M Arthur
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Demet Nalbant
- Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | - Henry A Feldman
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Bejan J Saeedi
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Jason Matthews
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Brian S Robinson
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Nourine A Kamili
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Ashley Bennett
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | | | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Rheinallt M Jones
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | | | - Andrew S Neish
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Ravi M Patel
- Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Peggy Nopoulos
- Department of Psychiatry, College of Medicine, University of Iowa, Iowa City, Iowa
| | - Michael K Georgieff
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - John D Roback
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - John A Widness
- Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | - Cassandra D Josephson
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia.,Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Sean R Stowell
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| |
Collapse
|
58
|
Singh P, Ochoa Allemant P, Brown J, Perides G, Freedman SD, Martin CR. Effect of polyunsaturated fatty acids on postnatal ileum development using the fat-1 transgenic mouse model. Pediatr Res 2019; 85:556-565. [PMID: 30653193 PMCID: PMC6397682 DOI: 10.1038/s41390-019-0284-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Long-chain polyunsaturated fatty acids (LCPUFAs) play a critical role in neonatal health. We hypothesized that LCPUFAs play an essential role in priming postnatal gut development. We studied the effect of LCPUFAs on postnatal gut development using fat-1 transgenic mice, which are capable of converting n-6 to n-3 LCPUFAs, and wild-type (WT) C57BL/6 mice. METHODS Distal ileum sections were collected from fat-1 and WT mice on days 3, 14, and 28. Fatty acid analyses, histology, RT-qPCR and intestinal permeability were performed. RESULTS Fat-1 mice, relative to WT mice, showed increased n-3 LCPUFAs levels (α-linolenic acid, docosahexaenoic acid, and eicosapentaenoic acid, p < 0.05) and decreased arachidonic acid levels (p < 0.05) in the ileum. Preweaning fat-1 mice, compared to WT, showed >50% reduced muc2, Tff3, TLR9, and Camp expression (p < 0.05), markers of the innate immune response. There was a >two-fold increased expression of Fzd5 and EphB2, markers of cell differentiation (p < 0.05), and Fabp2 and 6, regulators of fatty acid transport and metabolism (p < 0.05). Despite reduced expression of tight junction genes, intestinal permeability in fat-1 was comparable to WT mice. CONCLUSIONS Our data support the hypothesis that fatty acid profiles early in development modulate intestinal gene expression in formative domains, such as cell differentiation, tight junctions, other innate host defenses, and lipid metabolism.
Collapse
Affiliation(s)
- Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, MA,,Harvard Medical School, Boston, MA
| | - Pedro Ochoa Allemant
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, MA,,Harvard Medical School, Boston, MA
| | - Joanne Brown
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, MA,,Harvard Medical School, Boston, MA
| | - George Perides
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, MA,,Harvard Medical School, Boston, MA
| | - Steven D. Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, MA,,Division of Translational Research, Beth Israel Deaconess Medical Center, MA,,Harvard Medical School, Boston, MA
| | - Camilia R Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
59
|
Managlia E, Liu SXL, Yan X, Tan XD, Chou PM, Barrett TA, De Plaen IG. Blocking NF-κB Activation in Ly6c + Monocytes Attenuates Necrotizing Enterocolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:604-618. [PMID: 30593820 PMCID: PMC6412404 DOI: 10.1016/j.ajpath.2018.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease affecting premature infants with intestinal inflammation and necrosis. The neonatal intestinal inflammatory response is rich in macrophages, and blood monocyte count is low in human NEC. We previously found that NF-κB mediates the intestinal injury in experimental NEC. However, the role of NF-κB in myeloid cells during NEC remains unclear. Herein, inhibitor of kappaB kinase β (IKKβ), a critical kinase mediating NF-κB activation, was deleted in lysozyme M (Lysm)-expressing cells, which were found to be Cd11b+Ly6c+ monocytes but not Cd11b+Ly6c- macrophages in the dam-fed neonatal mouse intestine. NEC induced differentiation of monocytes into intestinal macrophages and up-regulation of monocyte recruitment genes (eg, L-selectin) in the macrophage compartment in wild-type mice, but not in pups with IKKβ deletion in Lysm+ cells. Thus, NF-κB is required for NEC-induced monocyte activation, recruitment, and differentiation in neonatal intestines. Furthermore, pups with Lysm-IKKβ deletion had improved survival and decreased incidence of severe NEC compared with littermate controls. Decreased NEC severity was not associated with an improved intestinal barrier. In contrast, NEC was unabated in mice with IKKβ deletion in intestinal epithelial cells. Together, these data suggest that recruitment of Ly6c+ monocytes into the intestine, NF-κB activation in these cells, and differentiation of Ly6c+ monocytes into macrophages are critical cellular and molecular events in NEC development to promote disease.
Collapse
Affiliation(s)
- Elizabeth Managlia
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Shirley X L Liu
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Xiaocai Yan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Xiao-Di Tan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Gastroenterology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Pauline M Chou
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Terrence A Barrett
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Isabelle G De Plaen
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
60
|
Roy SK, Meng Q, Sadowitz BD, Kollisch-Singule M, Yepuri N, Satalin J, Gatto LA, Nieman GF, Cooney RN, Clark D. Enteral administration of bacteria fermented formula in newborn piglets: A high fidelity model for necrotizing enterocolitis (NEC). PLoS One 2018; 13:e0201172. [PMID: 30036384 PMCID: PMC6056052 DOI: 10.1371/journal.pone.0201172] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022] Open
Abstract
Objective To develop an animal model which replicates neonatal NEC and characterizes the importance of bacterial fermentation of formula and short chain fatty acids (SCFAs) in its pathogenesis. Background NEC is a severe form of intestinal inflammation in preterm neonates and current models do not reproduce the human condition. Methods Three groups of newborn piglets: Formula alone (FO), Bacteria alone (E.coli: BO) and E.coli-fermented formula (FF) were anesthetized, instrumented and underwent post-pyloric injection of formula, bacteria or fermented-formula. SCFA levels were measured by gas chromatography-mass spectrometry. At 6 h bowel appearance was assessed, histologic and molecular analysis of intestine were performed. Gut inflammation (p65 NF-κB, TLR4, TNF-α, IL-1β), apoptosis (cleaved caspase-3, BAX, apoptosis) and tight junction proteins (claudin-2, occludin) were measured. Results SCFAs were increased in FF. Small bowel from FF piglet’s demonstrated inflammation, coagulative necrosis and pneumatosis resembling human NEC. Histologic gut injury (injury score, mast cell activation) were increased by Bacteria, but more severe in FF piglets. Intestinal expression of p65 NF-κB, NF-κB activation, TNF-α and IL-1β were increased in BO and markedly increased in the FF group (P<0.05 vs. FO). Intestine from Bacteria piglets demonstrated increased apoptotic index, pro-apoptotic protein expression and decreased tight junction proteins. These changes were more severe in FF piglets. Conclusions Our piglet model demonstrates the findings of NEC in human neonates: systemic acidosis, intestinal inflammation, pneumatosis and portal venous gas. Bacteria alone can initiate intestinal inflammation, injury and apoptosis, but bacterial fermentation of formula generates SCFAs which contribute to the pathogenesis of NEC.
Collapse
Affiliation(s)
- Shreyas K. Roy
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Qinghe Meng
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Benjamin D. Sadowitz
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Michaela Kollisch-Singule
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Natesh Yepuri
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Joshua Satalin
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Louis A. Gatto
- SUNY Cortland, Department of Biology, Cortland, New York, United States of America
| | - Gary F. Nieman
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
| | - Robert N. Cooney
- SUNY Upstate Medical University, Department of Surgery, Syracuse, New York, United States of America
- * E-mail:
| | - David Clark
- Albany Medical Center, Department of Pediatrics, Albany, New York, United States of America
| |
Collapse
|
61
|
There is an association between disease location and gestational age at birth in newborns submitted to surgery due to necrotizing enterocolitis. J Pediatr (Rio J) 2018; 94:320-324. [PMID: 28859914 DOI: 10.1016/j.jped.2017.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/08/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To evaluate if there are differences regarding disease location and mortality of necrotizing enterocolitis, according to the gestational age at birth, in newborns submitted to surgery due to enterocolite. METHODS A historical cohort study of 198 newborns submitted to surgery due to necrotizing enterecolitis in a tertiary hospital, from November 1991 to December 2012. The newborns were divided into different categories according to gestational age (<30 weeks, 30-33 weeks and 6 days, 34-36 weeks and 6 days, and ≥37 weeks), and were followed for 60 days after surgery. The inclusion criterion was the presence of histological findings of necrotizing enterocolitis in the pathology. Patients with single intestinal perforation were excluded. RESULTS The jejunum was the most commonly affected site in extremely premature infants (p=0.01), whereas the ileum was the most commonly affected site in premature infants (p=0.002), and the colon in infants born at term (p<0.001). With the increasing gestational age, it was observed that intestinal involvement decreased for the ileum and the jejunum (decreasing from 45% to 0% and from 5% to 0%, respectively), with a progressive increase in colon involvement (0% to 84%). Total mortality rate was 45.5%, and no statistical difference was observed in the mortality at different gestational ages (p=0.287). CONCLUSIONS In newborns submitted to surgery due to necrotizing enterocolitis, the disease in extremely preterm infants was more common in the jejunum, whereas in preterm infants, the most affected site was the ileum, and in newborns born close to term, it was the colon. No difference in mortality was observed according to the gestational age at birth.
Collapse
|
62
|
There is an association between disease location and gestational age at birth in newborns submitted to surgery due to necrotizing enterocolitis. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2018. [DOI: 10.1016/j.jpedp.2017.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
63
|
Denning NL, Prince JM. Neonatal intestinal dysbiosis in necrotizing enterocolitis. Mol Med 2018; 24:4. [PMID: 30134786 PMCID: PMC6016883 DOI: 10.1186/s10020-018-0002-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022] Open
Abstract
Necrotizing Enterocolitis (NEC) is one of the most devastating gastrointestinal diseases in neonates, particularly among preterm infants in whom surgical NEC is the leading cause of morbidity. NEC pathophysiology occurs in the hyper-reactive milieu of the premature gut after bacterial colonization. The resultant activation of the TLR4 pathway appears to be a strongly contributing factor. Advancements in metagenomics may yield new clarity to the relationship between the neonatal intestinal microbiome and the development of NEC. After a century without effective directed treatments, microbiome manipulation offers a promising therapeutic target for the prevention and treatment of this devastating disease.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA. .,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.
| | - Jose M Prince
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA.,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.,Trauma Institute, Northwell Health System, Manhasset, NY, 11030, USA
| |
Collapse
|
64
|
Lu L, Claud EC. Intrauterine Inflammation, Epigenetics, and Microbiome Influences on Preterm Infant Health. CURRENT PATHOBIOLOGY REPORTS 2018; 6:15-21. [PMID: 29938128 PMCID: PMC5978889 DOI: 10.1007/s40139-018-0159-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Significant research reveals that the microbiome modulates perinatal and postnatal health. This review aims to examine mechanisms by which intrauterine infection, the epigenome, and microbiome specifically influence preterm infant health outcomes. RECENT FINDINGS Intrauterine infection is a primary cause of preterm birth and can cause alterations in gene expression and epigenetic programming as well as postnatal inflammatory responses in the offspring. Insights from our own studies demonstrate epigenetic modifications of TLRs associated with exposure to intrauterine inflammation, as well as a cross talk between host epigenome and microbiome. Lastly, the gut microbiome modulates maturation of inflammatory pathways, which influences the development of preterm infants. SUMMARY We present a unifying theme that preterm infant outcomes are associated with modulation of host immune and inflammatory responses, which are influenced by acute intrauterine infection, epigenetic, and microbiome factors.
Collapse
Affiliation(s)
- Lei Lu
- Department of Pediatrics/Neonatology, University of Chicago, 900 E 57th Street, Chicago, IL 60637 USA
| | - Erika C. Claud
- Department of Pediatrics/Neonatology, University of Chicago, 5143 Maryland Street, Chicago, IL 60637 USA
| |
Collapse
|
65
|
Kessler SP, Obery DR, Nickerson KP, Petrey AC, McDonald C, de la Motte CA. Multifunctional Role of 35 Kilodalton Hyaluronan in Promoting Defense of the Intestinal Epithelium. J Histochem Cytochem 2018; 66:273-287. [PMID: 29290146 DOI: 10.1369/0022155417746775] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelium plays a critical role in host defense against orally acquired pathogens. Dysregulation of this protective barrier is a primary driver of inflammatory bowel diseases (Crohn's and ulcerative colitis) and also infant gastrointestinal infections. Previously, our lab reported that hyaluronan (HA) isolated from human milk induces the expression of the antimicrobial peptide β-defensin in vivo and protects against Salmonella Typhimurium infection of epithelial cells in vitro. In addition, we demonstrated that commercially available 35 kDa size HA induces the expression of β-defensin, upregulates the expression of tight junction protein zonula occludens-1 (ZO-1), and attenuates murine Citrobacter rodentium infection in vivo. In this current study, we report that HA35 remains largely intact and biologically active during transit through the digestive tract where it directly induces β-defensin expression upon epithelial cell contact. We also demonstrate HA35 abrogation of murine Salmonella Typhimurium infection as well as downregulation of leaky tight junction protein claudin-2 expression. Taken together, we propose a dual role for HA in host innate immune defense at the epithelial cell surface, acting to induce antimicrobial peptide production and also block pathogen-induced leaky gut. HA35 is therefore a promising therapeutic in the defense against bacterially induced colitis in compromised adults and vulnerable newborns.
Collapse
Affiliation(s)
- Sean P Kessler
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Dana R Obery
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Kourtney P Nickerson
- Division of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Aaron C Petrey
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Christine McDonald
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Carol A de la Motte
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
66
|
Ren W, Wu J, Li L, Lu Y, Shao Y, Qi Y, Xu B, He Y, Hu Y. Glucagon-Like Peptide-2 Improve Intestinal Mucosal Barrier Function in Aged Rats. J Nutr Health Aging 2018; 22:731-738. [PMID: 29806863 DOI: 10.1007/s12603-018-1022-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucagon-like peptide-2 (GLP-2) plays a major role in repairing impaired intestinal mucosa, but its mechanism in the improvement of intestinal barrier function during the aging process remains unclear. In this study, 26-month-old male Sprague-Dawley rats were randomized to control group and GLP-2 group treated with a dose of 250 μg•kg-1•d-1 by intraperitoneal injection. After 14 days of treatment, intestinal mucosal morphometric changes were observed by light microscopy and transmission electron microscopy (TEM). Small intestinal permeability was evaluated by fluorescein isothiocyanate (FITC)-labeled dextran. The mRNA and protein expression of Zonula Occludens-1 (ZO-1), occludin, claudin-1 and the GLP-2 receptor (GLP-2R) were detected by Real-time PCR and Western blot. Our results showed that GLP-2 administration significantly improved the age-related atrophy of intestinal mucosa and villi and increased small intestinal permeability. The mRNA and protein expression of ZO-1and occludin in ileum were up regulated in the GLP-2-treated old rats. In addition, the serum GLP-2 levels were negatively correlated with small intestinal permeability measured by FITC-dextran levels (r=-0.610, P<0.01). Taking all these data together, it is concluded that GLP-2 improved small intestinal epithelial barrier function in aged rats mainly by facilitating intestinal mucosa growth, alleviating the increased small intestinal permeability and increasing ZO-1 and occludin expression. Our observations provide evidence for the clinical significance of GLP-2 in preventing the intestinal epithelial barrier dysfunction during aging.
Collapse
Affiliation(s)
- W Ren
- Yu Hu, Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China, E-mail: , Tel:+86-021-64041990-3766, Fax:+86-21-64035399
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Cox CM, Lu R, Salcin K, Wilson JM. The Endosomal Protein Endotubin Is Required for Enterocyte Differentiation. Cell Mol Gastroenterol Hepatol 2017; 5:145-156. [PMID: 29322087 PMCID: PMC5756061 DOI: 10.1016/j.jcmgh.2017.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS During late embryonic development and through weaning, enterocytes of the ileum are highly endocytic. Defects in endocytosis and trafficking are implicated in neonatal disease, however, the mechanisms regulating trafficking during the developmental period are incompletely understood. The apical endosomal protein endotubin (EDTB) is highly expressed in the late embryonic and neonatal ileum. In epithelial cells in vitro, EDTB regulates both trafficking of tight junction proteins and proliferation through modulation of YAP activity. However, EDTB function during the endocytic stage of development of the intestine is unknown. METHODS By using Villin-CreERT2, we induced knockout of EDTB during late gestation and analyzed the impact on endocytic compartments and enterocyte structure in neonates using immunofluorescence, immunocytochemistry, and transmission electron microscopy. RESULTS Deletion of the apical endosomal protein EDTB in the small intestine during development impairs enterocyte morphogenesis, including loss of the apical endocytic complex, defective formation of the lysosomal compartment, and some cells had large microvillus-rich inclusions similar to those observed in microvillus inclusion disease. There also was a decrease in apical endocytosis and mislocalization of proteins involved in apical trafficking. CONCLUSIONS Our results show that EDTB-mediated trafficking within the epithelial cells of the developing ileum is important for maintenance of endocytic compartments and enterocyte integrity during early stages of gut development.
Collapse
Key Words
- AEC, apical endocytic complex
- AP, alkaline phosphatase
- CRISPR/Cas9, clustered regularly interspaced short palindromic repeats/cas9 endonuclease
- EDTB, endotubin
- EEA1, early endosomal antigen 1
- Endosomes
- Endotubin
- G, guide
- GFP, green fluorescent protein
- GTPase, guanosine triphosphatase
- KO, knockout
- LAMP1, lysosome-associated membrane protein 1
- MAMDC4, MAM domain containing 4
- MVID, microvillus inclusion disease
- P, postnatal day
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- Rab
- SDS, sodium dodecyl sulfate
- TBST, tris-buffered saline with 0.1% tween-20
- TEM, transmission electron microscopic
- TJ, tight junction
- Tight Junction
- Trafficking
Collapse
Affiliation(s)
- Christopher M. Cox
- Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Ruifeng Lu
- Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona,Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Kaan Salcin
- Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona,McGill University, Montreal, Canada
| | - Jean M. Wilson
- Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona,Correspondence Address correspondence to: Jean M. Wilson, PhD, Cell Biology and Anatomy, University of Arizona, PO Box 245044, Tucson, Arizona 85724. fax: (520) 626-2097.
| |
Collapse
|
68
|
Stewart AS, Pratt-Phillips S, Gonzalez LM. Alterations in Intestinal Permeability: The Role of the "Leaky Gut" in Health and Disease. J Equine Vet Sci 2017; 52:10-22. [PMID: 31000910 DOI: 10.1016/j.jevs.2017.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
All species, including horses, suffer from alterations that increase intestinal permeability. These alterations, also known as "leaky gut," may lead to severe disease as the normal intestinal barrier becomes compromised and can no longer protect against harmful luminal contents including microbial toxins and pathogens. Leaky gut results from a variety of conditions including physical stressors, decreased blood flow to the intestine, inflammatory disease, and pathogenic infections, among others. Several testing methods exist to diagnose these alterations in both a clinical and research setting. To date, most research has focused on regulation of the host immune response due to the wide variety of factors that can potentially influence the intestinal barrier. This article serves to review the normal intestinal barrier, measurement of barrier permeability, pathogenesis and main causes of altered permeability, and highlight potential alternative therapies of leaky gut in horses while relating what has been studied in other species. Conditions resulting in barrier dysfunction and leaky gut can be a major cause of decreased performance and also death in horses. A better understanding of the intestinal barrier in disease and ways to optimize the function of this barrier is vital to the long-term health and maintenance of these animals.
Collapse
Affiliation(s)
- Amy Stieler Stewart
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | | | - Liara M Gonzalez
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Center for Gastrointestinal Biology and Disease, Large Animal Models Core, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| |
Collapse
|