1
|
Bench to bedside - new insights into the pathogenesis of necrotizing enterocolitis. Nat Rev Gastroenterol Hepatol 2022; 19:468-479. [PMID: 35347256 DOI: 10.1038/s41575-022-00594-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 12/12/2022]
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of death and disability from gastrointestinal disease in premature infants. Recent discoveries have shed light on a unifying theorem to explain the pathogenesis of NEC, suggesting that specific treatments might finally be forthcoming. A variety of experiments have highlighted how the interaction between bacterial signalling receptors on the premature intestine and an abnormal gut microbiota incites a pro-inflammatory response in the intestinal mucosa and its underlying endothelium that leads to NEC. Central amongst the bacterial signalling receptors implicated in NEC development is the lipopolysaccharide receptor Toll-like receptor 4 (TLR4), which is expressed at higher levels in the premature gut than in the full-term gut. The high prenatal intestinal expression of TLR4 reflects the role of TLR4 in the regulation of normal gut development, and supports additional studies indicating that NEC develops in response to signalling events that occur in utero. This Review provides new evidence explaining the pathogenesis of NEC, explores new findings indicating that NEC development has origins before birth, and discusses future questions and opportunities for discovery in this field.
Collapse
|
2
|
Bell RL, Withers GS, Kuypers FA, Stehr W, Bhargava A. Stress and corticotropin releasing factor (CRF) promote necrotizing enterocolitis in a formula-fed neonatal rat model. PLoS One 2021; 16:e0246412. [PMID: 34111125 PMCID: PMC8191945 DOI: 10.1371/journal.pone.0246412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/25/2021] [Indexed: 11/18/2022] Open
Abstract
The etiology of necrotizing enterocolitis (NEC) is not known. Alterations in gut microbiome, mucosal barrier function, immune cell activation, and blood flow are characterized events in its development, with stress as a contributing factor. The hormone corticotropin-releasing factor (CRF) is a key mediator of stress responses and influences these aforementioned processes. CRF signaling is modulated by NEC's main risk factors of prematurity and formula feeding. Using an established neonatal rat model of NEC, we tested hypotheses that: (i) increased CRF levels-as seen during stress-promote NEC in formula-fed (FF) newborn rats, and (ii) antagonism of CRF action ameliorates NEC. Newborn pups were formula-fed to initiate gut inflammation and randomized to: no stress, no stress with subcutaneous CRF administration, stress (acute hypoxia followed by cold exposure-NEC model), or stress after pretreatment with the CRF peptide antagonist Astressin. Dam-fed unstressed and stressed littermates served as controls. NEC incidence and severity in the terminal ileum were determined using a histologic scoring system. Changes in CRF, CRF receptor (CRFRs), and toll-like receptor 4 (TLR4) expression levels were determined by immunofluorescence and immunoblotting, respectively. Stress exposure in FF neonates resulted in 40.0% NEC incidence, whereas exogenous CRF administration resulted in 51.7% NEC incidence compared to 8.7% in FF non-stressed neonates (p<0.001). Astressin prevented development of NEC in FF-stressed neonates (7.7% vs. 40.0%; p = 0.003). CRF and CRFR immunoreactivity increased in the ileum of neonates with NEC compared to dam-fed controls or FF unstressed pups. Immunoblotting confirmed increased TLR4 protein levels in FF stressed (NEC model) animals vs. controls, and Astressin treatment restored TLR4 to control levels. Peripheral CRF may serve as specific pharmacologic target for the prevention and treatment of NEC.
Collapse
MESH Headings
- Animals
- Female
- Rats
- Animals, Newborn
- Corticotropin-Releasing Hormone/metabolism
- Disease Models, Animal
- Enterocolitis, Necrotizing/metabolism
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/prevention & control
- Enterocolitis, Necrotizing/etiology
- Ileum/metabolism
- Ileum/pathology
- Peptide Fragments/metabolism
- Rats, Sprague-Dawley
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Stress, Physiological
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- Robert L. Bell
- East Bay Surgery Program, Department of Surgery, University of California San Francisco (UCSF) Benioff Children’s Hospital, Oakland, California, United States of America
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- The Permanente Medical Group, Department of Surgery, Walnut Creek, California, United States of America
| | - Ginger S. Withers
- Department of Biology, Whitman College, Walla Walla, Washington, United States of America
| | - Frans A. Kuypers
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- UCSF Benioff Children’s Hospital Oakland, Oakland, California, United States of America
| | - Wolfgang Stehr
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- UCSF Benioff Children’s Hospital Oakland, Oakland, California, United States of America
| | - Aditi Bhargava
- Department of Obstetrics and Gynecology, Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
3
|
Torikai M, Sugita K, Ibara S, Ishihara C, Kibe M, Murakami K, Shinyama S, Mukai M, Ikee T, Sueyoshi K, Noguchi H, Ieiri S. Prophylactic efficacy of enteral antifungal administration of miconazole for intestinal perforation, especially for necrotizing enterocolitis: a historical cohort study at a single institution. Surg Today 2020; 51:568-574. [PMID: 32886209 DOI: 10.1007/s00595-020-02125-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Despite improvements in neonatal intensive care, the outcomes of extremely-low-birth-weight infants (ELBWIs) with surgical diseases remain to be improved. We started administering enteral miconazole (MCZ) to ELBWIs from 2002 to prevent fungal infection. Since then, the incidence of intestinal perforation has significantly decreased. We investigated this prophylactic effect of MCZ against necrotizing enterocolitis (NEC) and focal intestinal perforation (FIP) and explored a new prophylactic concept against intestinal perforation. METHODS We designed a historical cohort study to evaluate the effect of MCZ for intestinal perforation in ELBWIs who underwent treatment in our neonatal intensive-care unit between January 1998 and December 2005. We divided these cases into two groups: the Pre-MCZ group and the Post-MCZ group. We compared the morbidity, clinical outcomes and pathological features of NEC and FIP. RESULTS The rate of intestinal perforation with NEC was significantly reduced after the introduction of MCZ (p = 0.007, odds ratio; 3.782, 95% confidence interval; 1.368-12.08). The pathological findings of NEC specimens showed that the accumulation of inflammatory cells was significantly reduced in the Post-MCZ group when compared with the Pre-MCZ group (p < 0.05). CONCLUSIONS The efficacy of the enteral administration of MCZ on intestinal perforation with NEC highlights a new prophylactic concept in the clinical management of ELBWIs.
Collapse
Affiliation(s)
- Motofumi Torikai
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.,Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Koshiro Sugita
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.,Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Satoshi Ibara
- Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Chie Ishihara
- Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Masaya Kibe
- Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Kenichi Murakami
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.,Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Shin Shinyama
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.,Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Motoi Mukai
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.,Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Takamasa Ikee
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.,Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Kazunobu Sueyoshi
- Department of Clinical Pathology, Kagoshima City Hospital, Kagoshima, Japan
| | - Hiroyuki Noguchi
- Department of Pediatric Surgery, Kagoshima City Hospital, Kagoshima, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.
| |
Collapse
|
4
|
He-Yang J, Zhang W, Liu J, Xue P, Zhou X. Human breast milk oligosaccharides attenuate necrotizing enterocolitis in rats by suppressing mast cell accumulation, DPPI activity and TLR4 expression in ileum tissue, and regulating mitochondrial damage of Caco-2 cells. Int Immunopharmacol 2020; 88:106881. [PMID: 32795899 DOI: 10.1016/j.intimp.2020.106881] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/09/2022]
Abstract
Necrotizing enterocolitis (NEC), a devastating infant disease characterized by severe intestinal necrosis, its pathogenesis is poorly understood, but appears to be multifactorial and highly associated with immaturity of gastrointestinal tract and immature innate-immune system. Breast-milk is effective strategy to protect infants against NEC. This study is using a NEC rat model to investigate the pathological mechanism of NEC involved intestinal-damages, and the therapeutic mechanism of sialylated human milk oligosaccharides (SHMOs) on NEC rats; also using cell model to investigate the effects of SHMOs on colon-epithelial cells (Caco-2) in-vitro. Extraction and characterization of SHMOs from breast milk, establishment of a NEC rat model, histopathological analysis and mast cell accounting of the terminal ileum were taken; The levels of DPPI, TLR4, IL-6, TNF-α, MMP-2/9 and glutathione were measured using various methods. Caco-2 cells were pre-treated with SHMOs and cultured with LPS, histamine, chymase or DPPI, cell viabilities and mitochondrial membrane potential were examined; flow cytometry was used to detect cell cycle. The accumulation of mast cells was found in the ileum of NEC rats, but prohibited by SHMOs treatment; the increased levels of TLR4, DPPI, IL-6, TNF-α, MMP-2/9 in NEC ileum were suppressed by SHMOs in-vivo. SHMOs prevented Caco-2 cells from LPS, histamine, chymase induced damages by surviving cell viability, regulating G0/G1 and S phase in cell cycles, and increasing mitochondrial membrane potential. These findings provide a new insight into the pharmacological mechanism of SHMOs treatment for NEC and suggest that SHMOs needs well attention for therapeutic aims.
Collapse
Affiliation(s)
- Jingqiu He-Yang
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China
| | - Wenting Zhang
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China; Department of Pharmacy, Affiliated Changzhou Children's Hospital of Nantong University, Yanling Road 468, Changzhou 213003, China
| | - Jie Liu
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China
| | - Peng Xue
- Department of Pharmacy, Affiliated Changzhou Children's Hospital of Nantong University, Yanling Road 468, Changzhou 213003, China
| | - Xiaoying Zhou
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China; The School of Medicine, the University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
5
|
Peterslund P, Rasmussen L, Qvist N, Hansen TP, Husby S, Detlefsen S. Frequencies of Immune Cells in the Human Small Bowel During Normal Gestation and in Necrotizing Enterocolitis. Fetal Pediatr Pathol 2019; 38:153-166. [PMID: 30689475 DOI: 10.1080/15513815.2018.1561774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Only few studies have quantitated the frequencies of immune cells in the small bowel mucosa and submucosa during gestation. The aims of this study were to describe the frequencies of T and B cells, eosinophils and mast cells in the normal small bowel mucosa and submucosa (NSB) in relation to gestational age (GA) and in the uninvolved small bowel (USB) of premature newborns with necrotizing enterocolitis (NEC). METHODS We obtained 36 NSB specimens (GA 12-41 weeks) and 8 NEC-USB specimens (GA 24-32 weeks) from autopsies and surgeries and performed immunostaining for CD3, CD79a, BMK-13 and tryptase as well as the histochemical stains giemsa and toluidine blue. Qualitative histological evaluation and two different quantitative cell-samplings were performed using digital imaging analysis with both TissuemorphDP® and newCAST® software. Linear regression analysis was performed with cell frequency as the dependent variable and GA and USB as the independent variables. RESULTS In the NSB specimens, we found significant linear correlations between cell frequencies and GA for all examined cell types, though B cell frequencies reached a plateau midway through gestation. In the USB cases, submucosal mast cell frequencies were higher than in the NSB specimens, while T cell frequencies were lower. In USB of NEC patients, we found a significant increase of mast cells and a significant decrease of T cells compared to NSB. CONCLUSION Throughout gestation, we found an increase of all examined immune cell types in the normal small bowel, while the number of B cells came to a standstill at midway. Future studies should examine subtypes of T cells and also include histiocytes. A larger amount of small bowel specimens, covering the full gestational age, would be of great value.
Collapse
Affiliation(s)
- Paul Peterslund
- a Department of Pathology , Odense University Hospital , Odense , Denmark
| | - Lene Rasmussen
- b Department of Internal Medicine , Hospital Lillebaelt Kolding Hospital , Kolding , Denmark
| | - Niels Qvist
- c Department of Surgery , Odense University Hospital , Odense , Denmark
| | | | - Steffen Husby
- e Hans Christian Andersen Children's Hospital , Odense University Hospital , Odense , Denmark
| | - Sönke Detlefsen
- a Department of Pathology , Odense University Hospital , Odense , Denmark.,f Department of Clinical Research , Faculty of Health Sciences, University of Southern Denmark , Odense , Denmark
| |
Collapse
|
6
|
Neu J, Pammi M. Necrotizing enterocolitis: The intestinal microbiome, metabolome and inflammatory mediators. Semin Fetal Neonatal Med 2018; 23:400-405. [PMID: 30172660 DOI: 10.1016/j.siny.2018.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Necrotizing enterocolitis (NEC) is a disease of preterm infants and associated with significant mortality and morbidity. Although the pathogenesis of NEC is not clear, microbial dysbiosis, with a bloom of the phylum Proteobacteria, has been reported. Antibiotics and the use of H2 blockers, which affect the gut microbiome, are associated with increased incidence of NEC. In association with dysbiosis, inflammatory processes are upregulated with increased Toll-like receptor signaling, leading to translocation of nuclear factor kappa-β, a transcription factor that induces transcription of various pro-inflammatory cytokines and chemokines. Microbial metabolites, short chain fatty acids including acetate and butyrate, may modulate immunity, inflammation, intestinal integrity and regulate transcription by epigenetic mechanisms. Evaluation of microbiome and metabolome may provide biomarkers for early diagnosis of NEC and microbial therapeutic approaches to correct microbial dysbiosis.
Collapse
Affiliation(s)
- Josef Neu
- Section of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| | - Mohan Pammi
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
7
|
Intestinal microbiota development and gestational age in preterm neonates. Sci Rep 2018; 8:2453. [PMID: 29410448 PMCID: PMC5802739 DOI: 10.1038/s41598-018-20827-x] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/24/2018] [Indexed: 01/11/2023] Open
Abstract
The intestinal microbiota is an important contributor to the health of preterm infants, and may be destabilized by a number of environmental factors and treatment modalities. How to promote the development of a healthy microbiota in preterm infants is largely unknown. We collected fecal samples from 45 breastfed preterm very low birth weight (birth weight < 1500 g) infants from birth until 60 days postnatal age to characterize the intestinal microbiota development during the first weeks of life in preterm infants. Fecal microbiota composition was determined by 16S rRNA amplicon sequencing. The main driver of microbiota development was gestational age; antibiotic use had strong but temporary effects and birth mode had little influence. Microbiota development proceeded in four phases indicated by the dominance of Staphylococcus, Enterococcus, Enterobacter, and finally Bifidobacterium. The Enterococcus phase was only observed among the extremely premature infants and appeared to delay the microbiota succession. The results indicate that hospitalized preterm infants receiving breast milk may develop a normal microbiota resembling that of term infants.
Collapse
|
8
|
Ginzel M, Feng X, Kuebler JF, Klemann C, Yu Y, von Wasielewski R, Park JK, Hornef MW, Vieten G, Ure BM, Kaussen T, Gosemann JH, Mayer S, Suttkus A, Lacher M. Dextran sodium sulfate (DSS) induces necrotizing enterocolitis-like lesions in neonatal mice. PLoS One 2017; 12:e0182732. [PMID: 28817583 PMCID: PMC5560643 DOI: 10.1371/journal.pone.0182732] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is an inflammatory bowel disease of preterm human newborns with yet unresolved etiology. An established neonatal murine model for NEC employs oral administration of lipopolysaccharides (LPS) combined with hypoxia/hypothermia. In adult mice, feeding dextran sodium sulfate (DSS) represents a well-established model for experimental inflammatory bowel disease. Here we investigated the effect of DSS administration on the neonatal murine intestine in comparison with the established NEC model. Methods 3-day-old C57BL/6J mice were either fed formula containing DSS or LPS. LPS treated animals were additionally stressed by hypoxia/hypothermia twice daily. After 72 h, mice were euthanized, their intestinal tissue harvested and analyzed by histology, qRT-PCR and flow cytometry. For comparison, adult C57BL/6J mice were fed with DSS for 8 days and examined likewise. Untreated, age matched animals served as controls. Results Adult mice treated with DSS exhibited colonic inflammation with significantly increased Cxcl2 mRNA expression. In contrast, tissue inflammation in neonatal mice treated with DSS or LPS plus hypoxia/hypothermia was present in colon and small intestine as well. Comparative analysis of neonatal mice revealed a significantly increased lesion size and intestinal Cxcl2 mRNA expression after DSS exposure. Whereas LPS administration mainly induced local neutrophil recruitment, DSS treated animals displayed increased monocytes/macrophages infiltration. Conclusions Our study demonstrates the potential of DSS to induce NEC-like lesions accompanied by a significant humoral and cellular immune response in the small and large intestine of neonatal mice. The new model therefore represents a good alternative to LPS plus hypoxia/hypothermia administration requiring no additional physical stress.
Collapse
Affiliation(s)
- Marco Ginzel
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Xiaoyan Feng
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | | | - Christian Klemann
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Yi Yu
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | | | - Joon-Keun Park
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | | | - Gertrud Vieten
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Benno M. Ure
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Torsten Kaussen
- Department of Pediatric Cardiology and Pediatric Intensive Care, Hannover, Germany
| | | | - Steffi Mayer
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Anne Suttkus
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
9
|
Ferretti E, Tremblay E, Thibault MP, Grynspan D, Burghardt KM, Bettolli M, Babakissa C, Levy E, Beaulieu JF. The nitric oxide synthase 2 pathway is targeted by both pro- and anti-inflammatory treatments in the immature human intestine. Nitric Oxide 2017; 66:53-61. [PMID: 28315470 DOI: 10.1016/j.niox.2017.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/16/2017] [Accepted: 03/13/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM NO synthase 2 (NOS2) was recently identified as one the most overexpressed genes in intestinal samples of premature infants with necrotizing enterocolitis (NEC). NOS2 is widely implicated in the processes of epithelial cell injury/apoptosis and host immune defense but its specific role in inflammation of the immature human intestinal mucosa remains unclear. Interestingly, factors that prevent NEC such as epidermal growth factor (EGF) attenuate the inflammatory response in the mid-gestation human small intestine using serum-free organ culture while drugs that are associated with NEC occurrence such as the non-steroidal anti-inflammatory drug, indomethacin (INDO), exert multiple detrimental effects on the immature human intestine. In this study we investigate the potential role of NOS2 in modulating the gut inflammatory response under protective and stressful conditions by determining the expression profile of NOS2 and its downstream pathways in the immature intestine. METHODS Gene expression profiles of cultured mid-gestation human intestinal explants were investigated in the absence or presence of a physiological concentration of EGF (50 ng/ml) or 1 μM INDO for 48 h using Illumina whole genome microarrays, Ingenuity Pathway Analysis software and quantitative PCR to investigate the expression of NOS2 and NOS2-pathway related genes. RESULTS In the immature intestine, NOS2 expression was found to be increased by EGF and repressed by INDO. Bioinformatic analysis identified differentially regulated pathways where NOS2 is known to play an important role including citrulline/arginine metabolism, epithelial cell junctions and oxidative stress. At the individual gene level, we identified many differentially expressed genes of the citrulline/arginine metabolism pathway such as ARG1, ARG2, GLS, OAT and OTC in response to EGF and INDO. Gene expression of tight junction components such as CLDN1, CLDN2, CLDN7 and OCN and of antioxidant markers such as DUOX2, GPX2, SOD2 were also found to be differentially modulated by EGF and INDO. CONCLUSION These results suggest that the protective effect of EGF and the deleterious influence of INDO on the immature intestine could be mediated via regulation of NOS2. Pathways downstream of NOS2 involved with these effects include metabolism linked to NO production, epithelial barrier permeability and antioxidant expression. These results suggest that NOS2 is a likely regulator of the inflammatory response in the immature human gut and may provide a mechanistic basis for the protective effect of EGF and the deleterious effects of INDO.
Collapse
Affiliation(s)
- Emanuela Ferretti
- Research Consortium on Child Intestinal Inflammation, Division of Neonatology, Department of Pediatrics, University of Ottawa, Ottawa, Canada
| | - Eric Tremblay
- Research Consortium on Child Intestinal Inflammation, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Marie-Pier Thibault
- Research Consortium on Child Intestinal Inflammation, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - David Grynspan
- Research Consortium on Child Intestinal Inflammation, Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Canada
| | - Karolina M Burghardt
- Research Consortium on Child Intestinal Inflammation, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children and Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Marcos Bettolli
- Research Consortium on Child Intestinal Inflammation, Department of Surgery, University of Ottawa, Ottawa, Canada
| | - Corentin Babakissa
- Research Consortium on Child Intestinal Inflammation, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Emile Levy
- Research Consortium on Child Intestinal Inflammation, Department of Nutrition, CHU Sainte-Justine, Université de Montréal, Montréal, Canada
| | - Jean-François Beaulieu
- Research Consortium on Child Intestinal Inflammation, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada.
| |
Collapse
|
10
|
Torikai M, Ibara S, Ieiri S, Hamada T, Noguchi H, Sueyoshi K, Fukuda T, Abeyama K. Prophylactic efficacy of enteral miconazole administration for neonatal intestinal perforation and its potential mechanism. Pediatr Surg Int 2016; 32:953-7. [PMID: 27473010 DOI: 10.1007/s00383-016-3946-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE Intestinal perforation (IP) is a fatal complication in extremely low birth weight infants (ELBWI). We started administrating enteral miconazole (MCZ) to ELBWI in 2002. Since then, the incidence of IP has significantly decreased. The aim of this study was to elucidate the prophylactic effect of MCZ for the treatment of neonatal IP, and to establish a new prophylactic concept for this disease. METHODS In in vivo experiments, the effects of MCZ were examined histopathologically using a mouse model of intestinal ischemia. In in vitro experiments, the cytoprotective effect of MCZ against hypoxia was evaluated using Caco-2 intestinal cells, and its anti-inflammatory potential using a co-culture model of Caco-2 and HL60 cells. RESULTS MCZ showed a tissue protective effect against intestinal ischemia. MCZ reduced high mobility group-box 1 (HMGB1) release in Caco-2 cells under hypoxic stress and attenuated the potential to activate co-cultured HL60 leukocytes with Caco-2 cells by suppressing interleukin-8 (IL-8). CONCLUSION MCZ may have preventive roles in the clinical management of IP in ELBWI by the suppression of IL-8 and HMGB-1.
Collapse
Affiliation(s)
- Motofumi Torikai
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Satoshi Ibara
- Department of Neonatology, Perinatal Medical Center, Kagoshima City Hospital, Kagoshima, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Takashi Hamada
- Department of Preventive Medicine supported by Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| | - Hiroyuki Noguchi
- Division of Pediatric Surgery, Kagoshima City Hospital, Kagoshima, Japan
| | - Kazunobu Sueyoshi
- Division of Clinical Pathology, Kagoshima City Hospital, Kagoshima, Japan
| | - Takeo Fukuda
- Department of Preventive Medicine supported by Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| | - Kazuhiro Abeyama
- Department of Preventive Medicine supported by Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
11
|
Warner BB, Deych E, Zhou Y, Hall-Moore C, Weinstock GM, Sodergren E, Shaikh N, Hoffmann JA, Linneman LA, Hamvas A, Khanna G, Rouggly-Nickless LC, Ndao IM, Shands BA, Escobedo M, Sullivan JE, Radmacher PG, Shannon WD, Tarr PI. Gut bacteria dysbiosis and necrotising enterocolitis in very low birthweight infants: a prospective case-control study. Lancet 2016; 387:1928-36. [PMID: 26969089 PMCID: PMC5553277 DOI: 10.1016/s0140-6736(16)00081-7] [Citation(s) in RCA: 300] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Gut bacteria might predispose to or protect from necrotising enterocolitis, a severe illness linked to prematurity. In this observational prospective study we aimed to assess whether one or more bacterial taxa in the gut differ between infants who subsequently develop necrotising enterocolitis (cases) and those who do not (controls). METHODS We enrolled very low birthweight (1500 g and lower) infants in the primary cohort (St Louis Children's Hospital) between July 7, 2009, and Sept 16, 2013, and in the secondary cohorts (Kosair Children's Hospital and Children's Hospital at Oklahoma University) between Sept 12, 2011 and May 25, 2013. We prospectively collected and then froze stool samples for all infants. Cases were defined as infants whose clinical courses were consistent with necrotising enterocolitis and whose radiographs fulfilled criteria for Bell's stage 2 or 3 necrotising enterocolitis. Control infants (one to four per case; not fixed ratios) with similar gestational ages, birthweight, and birth dates were selected from the population after cases were identified. Using primers specific for bacterial 16S rRNA genes, we amplified and then pyrosequenced faecal DNA from stool samples. With use of Dirichlet multinomial analysis and mixed models to account for repeated measures, we identified host factors, including development of necrotising enterocolitis, associated with gut bacterial populations. FINDINGS We studied 2492 stool samples from 122 infants in the primary cohort, of whom 28 developed necrotising enterocolitis; 94 infants were used as controls. The microbial community structure in case stools differed significantly from those in control stools. These differences emerged only after the first month of age. In mixed models, the time-by-necrotising-enterocolitis interaction was positively associated with Gammaproteobacteria (p=0·0010) and negatively associated with strictly anaerobic bacteria, especially Negativicutes (p=0·0019). We studied 1094 stool samples from 44 infants in the secondary cohorts. 18 infants developed necrotising enterocolitis (cases) and 26 were controls. After combining data from all cohorts (166 infants, 3586 stools, 46 cases of necrotising enterocolitis), there were increased proportions of Gammaproteobacteria (p=0·0011) and lower proportions of both Negativicutes (p=0·0013) and the combined Clostridia-Negativicutes class (p=0·0051) in infants who went on to develop necrotising enterocolitis compared with controls. These associations were strongest in both the primary cohort and the overall cohort for infants born at less than 27 weeks' gestation. INTERPRETATION A relative abundance of Gammaproteobacteria (ie, Gram-negative facultative bacilli) and relative paucity of strict anaerobic bacteria (especially Negativicutes) precede necrotising enterocolitis in very low birthweight infants. These data offer candidate targets for interventions to prevent necrotising enterocolitis, at least among infants born at less than 27 weeks' gestation. FUNDING National Institutes of Health (NIH), Foundation for the NIH, the Children's Discovery Institute.
Collapse
Affiliation(s)
- Barbara B Warner
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Elena Deych
- Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Yanjiao Zhou
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - George M Weinstock
- McDonnell Genome Institute, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Erica Sodergren
- McDonnell Genome Institute, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Julie A Hoffmann
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Laura A Linneman
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Aaron Hamvas
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Geetika Khanna
- Department of Radiology, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | | | - I Malick Ndao
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Berkley A Shands
- Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Marilyn Escobedo
- Department of Pediatrics, University of Oklahoma School of Medicine, Oklahoma City, OK, USA
| | - Janice E Sullivan
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Paula G Radmacher
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - William D Shannon
- Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA.
| | - Phillip I Tarr
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA; Department of Molecular Microbiology, Washington University in St Louis School of Medicine, St Louis, MO, USA.
| |
Collapse
|
12
|
Application of Laser Capture Microdissection and 16S rRNA Gene Polymerase Chain Reaction in the Analysis of Bacteria Colonizing the Intestinal Tissue of Neonates With Necrotizing Enterocolitis. Pediatr Infect Dis J 2015; 34:e279-89. [PMID: 26372849 DOI: 10.1097/inf.0000000000000837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in newborns. However, the pathogenesis of NEC remains unclear because most bacterial characterizations of alleged pathogens have been performed via the analysis of human fecal samples and experimental animal studies. The objective is to investigate the microbial composition of NEC using inflamed intestinal tissue surgically removed from neonates diagnosed with NEC (n = 18). METHODS We obtained intestinal tissues via a combination of laser capture microdissection and Gram staining, which was used to mark individual bacteria. Tissues with congenital intestinal atresia (n = 7) served as control specimens. An analysis of the 16S rRNA of each sample was performed via polymerase chain reaction-denaturing gradient gel electrophoresis. RESULTS Numerous bacteria were observed in the inflamed intestinal wall tissue samples obtained from neonates with NEC following Gram staining and examination under an optical microscope. The total number of types detected by polymerase chain reaction-denaturing gradient gel electrophoresis was 12.17 ± 2.83 per infant with NEC, whereas only 2.57 ± 1.81 types were detected in each infant with congenital intestinal atresia. Proteobacteria had the highest constituent ratio (188 of 285) of all detected clone sequences in the NEC group. Additionally, Pseudomonas sp., Acinetobacter sp., Klebsiella sp., Clostridium sp., Ochrobactrum sp. and Arcobacter sp. were detected only in the NEC group. CONCLUSIONS The combination of Gram staining and laser capture microdissection was a reliable method to obtain and prepare tissue samples for processing. NEC was associated with multiple species of bacteria, and microflora within the disease-affected sites may be relatively specific and stable. Proteobacteria demonstrated the highest constituent ratio. Our observations warrant closer examination of the 6 bacterial genera that were only detected in NEC, particularly Clostridium sp., which may be closely correlated with pneumatosis intestinalis.
Collapse
|
13
|
Fisher JG, Bairdain S, Sparks EA, Khan FA, Archer JM, Kenny M, Edwards EM, Soll RF, Modi BP, Yeager S, Horbar JD, Jaksic T. Serious Congenital Heart Disease and Necrotizing Enterocolitis in Very Low Birth Weight Neonates. J Am Coll Surg 2015; 220:1018-1026.e14. [DOI: 10.1016/j.jamcollsurg.2014.11.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/26/2014] [Indexed: 11/17/2022]
|
14
|
Fulde M, Hornef MW. Maturation of the enteric mucosal innate immune system during the postnatal period. Immunol Rev 2015; 260:21-34. [PMID: 24942679 DOI: 10.1111/imr.12190] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The innate immune system instructs the host on microbial exposure and infection. This information is critical to mount a protective innate and adaptive host response to microbial challenge, but is also involved in homeostatic and adaptive processes that adjust the organism to meet environmental requirements. This is of particular importance for the neonatal host during the transition from the protected fetal life to the intense and dynamic postnatal interaction with commensal and pathogenic microorganisms. Here, we discuss both adaptive and developmental mechanisms of the mucosal innate immune system that prevent inappropriate stimulation and facilitate establishment of a stable homeostatic host-microbial interaction after birth.
Collapse
Affiliation(s)
- Marcus Fulde
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
15
|
Reisinger KW, Elst M, Derikx JPM, Nikkels PGJ, de Vries B, Adriaanse MPM, Jellema RK, Kramer BWW, Wolfs TGAM. Intestinal fatty acid-binding protein: a possible marker for gut maturation. Pediatr Res 2014; 76:261-8. [PMID: 24956227 DOI: 10.1038/pr.2014.89] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 04/02/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Gut immaturity is linked with postnatal intestinal disorders. However, biomarkers to assess the intestinal developmental stage around birth are lacking. The aim of this study was to gain more insight on intestinal fatty acid-binding protein (I-FABP) as an indicator of gut maturity. METHODS Antenatal I-FABP distribution and release was investigated in extremely premature, moderately premature, and term lambs, and these findings were verified in human urinary samples. Ileal I-FABP distribution was confirmed in autopsy material within 24 h postnatally. RESULTS Median (range) serum I-FABP levels were lower in extremely premature lambs compared with moderately premature lambs (156 (50.0-427) vs. 385 (100-1,387) pg/ml; P = 0.02). Contrarily, median early postnatal urine I-FABP levels in human infants were higher in extremely premature compared with moderately premature and term neonates (1,219 (203-15,044) vs. 256 (50-1,453) and 328 (96-1,749) pg/ml; P = 0.008 and P = 0.04, respectively). I-FABP expression was most prominent in nonvacuolated enterocytes and increased with rising gestational age (GA) in ovine and human tissue samples. The epithelial distribution pattern changed from a phenotype displaying I-FABP-positive enterocytes merely in the crypts early in gestation into a phenotype with I-FABP expressing cells exclusively present in the villus tips at term in ovine and human tissue. CONCLUSION In this ovine and human study, increasing GA is accompanied by an increase in I-FABP tissue content. Cord I-FABP levels correlate with gestation in ovine fetuses, identifying I-FABP as a marker for gut maturation. Raised postnatal urine I-FABP levels in preterm human infants may indicate intestinal injury and/or inflammation in utero.
Collapse
Affiliation(s)
- Kostan W Reisinger
- Department of Surgery, Maastricht University Medical Center and Nutrition and Toxicology Research Institute (NUTRIM), Maastricht, The Netherlands
| | - Marieke Elst
- Department of Pediatrics, Maastricht University Medical Center and School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Joep P M Derikx
- Department of Surgery, Maastricht University Medical Center and Nutrition and Toxicology Research Institute (NUTRIM), Maastricht, The Netherlands
| | - Peter G J Nikkels
- Department of Pathology, University Medical Center, Utrecht, The Netherlands
| | - Bart de Vries
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marlou P M Adriaanse
- Department of Pediatrics & Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Reint K Jellema
- 1] Department of Pediatrics, Maastricht University Medical Center and School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands [2] Department of Pediatrics & School for Mental Health and NeuroScience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Boris W W Kramer
- 1] Department of Pediatrics, Maastricht University Medical Center and School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands [2] Department of Pediatrics & School for Mental Health and NeuroScience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Center and School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| |
Collapse
|
16
|
Abstract
BACKGROUND Several risk factors have been implicated in the development of necrotising enterocolitis (NEC). However, little has been reported on the risk factors for NEC in infants born in a setting where exposure to maternal human immunodeficiency virus (HIV) is prevalent. OBJECTIVES To determine maternal and infant characteristics associated with NEC in a setting with a high prevalence of HIV infection and to compare clinical presentation and mortality of NEC in HIV-exposed and unexposed infants. METHODS This was a retrospective, case-control study. Infants with a confirmed diagnosis of NEC, admitted between January 2005 and December 2008 were identified as cases. Two controls for each case were selected by matching them for birthweight, gestational age and date of birth. Hospital records were reviewed for maternal and infant characteristics. RESULTS One hundred and ten infants with confirmed NEC were identified and 220 were selected as controls. Median birthweight and gestational age were similar between cases and controls, (1370 and 1380 g, P = 0·96, and 31 weeks each, P = 0·62, respectively). Lack of use of antenatal corticosteroids (ANS) (OR 2·77, 95% CI 1·42-5·38, P = 0·003), presence of chorio-amnionitis (OR 7·28, 95% CI 2·16-24·51, P = 0·001) and not mechanically ventilated at birth (OR 3·54, 95% CI 1·29-9·69, P = 0·01) were independently associated with NEC. Maternal HIV status was not associated with NEC. Clinical presentation and mortality from NEC were similar between HIV-exposed and unexposed infants. CONCLUSIONS Infants who develop NEC were less likely to have received mechanical ventilation at birth, suggesting that they were less critically ill when born. Use of ANS should be encouraged as it is protective against NEC. Infants born to HIV-infected mothers were not at increased risk of NEC.
Collapse
|
17
|
Jensen ML, Thymann T, Cilieborg MS, Lykke M, Mølbak L, Jensen BB, Schmidt M, Kelly D, Mulder I, Burrin DG, Sangild PT. Antibiotics modulate intestinal immunity and prevent necrotizing enterocolitis in preterm neonatal piglets. Am J Physiol Gastrointest Liver Physiol 2014; 306:G59-71. [PMID: 24157972 PMCID: PMC4073901 DOI: 10.1152/ajpgi.00213.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/11/2013] [Indexed: 01/31/2023]
Abstract
Preterm birth, bacterial colonization, and formula feeding predispose to necrotizing enterocolitis (NEC). Antibiotics are commonly administered to prevent sepsis in preterm infants, but it is not known whether this affects intestinal immunity and NEC resistance. We hypothesized that broad-spectrum antibiotic treatment improves NEC resistance and intestinal structure, function, and immunity in neonates. Caesarean-delivered preterm pigs were fed 3 days of parenteral nutrition followed by 2 days of enteral formula. Immediately after birth, they were assigned to receive either antibiotics (oral and parenteral doses of gentamycin, ampicillin, and metronidazole, ANTI, n = 11) or saline in the control group (CON, n = 13), given twice daily. NEC lesions and intestinal structure, function, microbiology, and immunity markers were recorded. None of the ANTI but 85% of the CON pigs developed NEC lesions by day 5 (0/11 vs. 11/13, P < 0.05). ANTI pigs had higher intestinal villi (+60%), digestive enzyme activities (+53-73%), and goblet cell densities (+110%) and lower myeloperoxidase (-51%) and colonic microbial density (10(5) vs. 10(10) colony-forming units, all P < 0.05). Microarray transcriptomics showed strong downregulation of genes related to inflammation and innate immune response to microbiota and marked upregulation of genes related to amino acid metabolism, in particular threonine, glucose transport systems, and cell cycle in 5-day-old ANTI pigs. In a follow-up experiment, 5 days of antibiotics prevented NEC at least until day 10. Neonatal prophylactic antibiotics effectively reduced gut bacterial load, prevented NEC, intestinal atrophy, dysfunction, and inflammation and enhanced expression of genes related to gut metabolism and immunity in preterm pigs.
Collapse
Affiliation(s)
- Michael L Jensen
- Dept. of Nutrition, Exercise and Sports, Faculty of Science, Univ. of Copenhagen, 30 Rolighedsvej, DK-1958 Frederiksberg C,.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Prevention of necrotizing enterocolitis in preterm very low birth weight infants: is it feasible? J Formos Med Assoc 2013; 113:490-7. [PMID: 23701837 DOI: 10.1016/j.jfma.2013.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/08/2013] [Accepted: 03/29/2013] [Indexed: 01/11/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is still one of the most catastrophic intestinal emergencies in preterm very low-birth weight infants. Primary prevention of NEC should be the priority, since NEC frequently progresses from nonspecific signs, to extensive necrosis within a matter of hours with medical or surgical treatment, making successful treatment and secondary prevention difficult to achieve. Currently available strategies for primary prevention of NEC include antenatal glucocorticosteroids, breast milk feeding, cautious feeding strategy, fluid restriction and probiotics. Nonetheless, based on current research evidence, mixed flora probiotics, and/or breast milk feeding, would appear to be the most effective feasible methods in the prevention of NEC at present.
Collapse
|
19
|
Abstract
Necrotizing enterocolitis (NEC) primarily affects premature infants. It is less common in term and late preterm infants. The age of onset is inversely related to the postmenstrual age at birth. In term infants, NEC is commonly associated with congenital heart diseases. NEC has also been associated with other anomalies. More than 85% of all NEC cases occur in very low birth weight infants or in very premature infants. Despite incremental advances in our understanding of the clinical presentation and pathophysiology of NEC, universal prevention of this disease continues to elude us even in the twenty-first century.
Collapse
MESH Headings
- Age of Onset
- Disease Management
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/epidemiology
- Enterocolitis, Necrotizing/therapy
- Humans
- Infant, Newborn
- Infant, Premature/physiology
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/epidemiology
- Infant, Premature, Diseases/therapy
- Intestinal Perforation/diagnosis
- Intestines/diagnostic imaging
- Intestines/microbiology
- Intestines/physiopathology
- Pneumoperitoneum/diagnosis
- Prevalence
- Radiography
- Risk
Collapse
Affiliation(s)
- Renu Sharma
- Division of Neonatology, Department of Pediatrics, University of Florida College of Medicine at Jacksonville, 655 West 8th Street, Jacksonville, FL 32209, USA.
| | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Bacterial colonization of the infant intestinal tract begins at birth. We are at the forefront of understanding complex relationships between bacteria and multiple parameters of health of the developing infant. Moreover, the establishment of the microbiome in the critical neonatal period is potentially foundational for lifelong health and disease susceptibility. Recent studies utilizing state-of-the-art culture-independent technologies have begun to increase our knowledge about the gut microbiome in infancy, the impact of multiple exposures, and its effects on immune response and clinical outcomes such as allergy and infection. RECENT FINDINGS Postnatal exposures play a central role in the complex interactions between the nearly blank canvas of the neonatal intestine, whereas genetic factors do not appear to be a major factor. Infant microbial colonization is affected by delivery mode, dietary exposures, antibiotic exposure, and environmental toxicants. Successive microbiome acquisition in infancy is likely a determinant of early immune programming, subsequent infection, and allergy risk. SUMMARY The novel investigation of the neonatal microbiome is beginning to unearth substantial information, with a focus on immune programming that coevolves with the developing microbiome early in life. Several exposures common to neonatal and infant populations could exert pressure on the development of the microbiome and major diseases including allergy and infection in large populations.
Collapse
|
21
|
Abstract
TRAGI (transfusion-related acute gut injury) is an acronym we proposed to characterize a severe neonatal gastrointestinal reaction temporally related to a transfusion of packed blood red cells (PRBCs) for anemia in very low birth weights. The following are in support of a causative relationship: (1) the timing of necrotizing enterocolitis after a PRBC transfusion not being random, (2) traditional risk factors for necrotizing enterocolitis are often absent, (3) significant anemia appears to be a universal finding, (4) the age of donor blood is often slightly older than controls, (5) TRAGI is not postnatal age dependent, and (6) TRAGI does not show a centering at 31 weeks' postconceptual age as does nontransfusion-related NEC. Although TRAGI is linked to the timing of PRBC transfusions, we propose a novel hypothesis that the convergence at 31 weeks' postconceptual age for classic NEC approximates the age of presentation of other oxygen delivery and neovascularization syndromes (eg, retinopathy of prematurity), suggesting its etiologic link to a generalized systemic maturational mechanism or another common developmental theme. This report will begin by reviewing the history of the clinical presentation and discovery of TRAGI and will then analyze various pathophysiologic mechanisms that may account for the phenomenon when clinicians render therapies. We will end by a call to action for randomized clinical trials to test various etiologic theories.
Collapse
Affiliation(s)
- Edmund F La Gamma
- The Regional Neonatal Intensive Care Unit, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY 10595, USA.
| | | |
Collapse
|
22
|
Association between mannose-binding lectin gene polymorphisms and necrotizing enterocolitis in preterm infants. J Pediatr Gastroenterol Nutr 2012; 55:160-5. [PMID: 22331020 DOI: 10.1097/mpg.0b013e31824e5f7a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The aim of the present study was to evaluate whether polymorphisms of the mannose-binding lectin (MBL-2) gene and MBL serum levels on admission to neonatal intensive care unit are associated with necrotizing enterocolitis (NEC) in preterm infants and to verify MBL expression in NEC bowels. METHODS In this retrospective cohort study, 107 neonates (41 with NEC and 66 controls) were included. MBL-2 genotyping for the promoter polymorphism -221 and for the exon 1 variant alleles at codons 52, 54, and 57 was performed. MBL levels were determined by enzyme-linked immunosorbent assay in 55 infants. Immunohistochemical staining for MBL expression was performed on bowel specimens. The main study outcome was severe NEC (Bell stages II/III). RESULTS The -221 Y allele and the MBL-2 YY genotype were more frequent in neonates with severe NEC than in controls (P = 0.04 and P = 0.004, respectively). In the multivariate analysis, the MBL-2 YA/YA genotype was associated with NEC (odds ratio = 3.03, 95% confidence interval 1.13%-8.13%, P = 0.024). Neonates with NEC had MBL level on admission >400 ng/mL more frequently than controls (P = 0.043). Among neonates with severe NEC, the deceased neonates were carriers of high or intermediate producing MBL-2 genotypes (P = 0.035). Finally, MBL was highly expressed in intestinal tissue from infants with NEC. CONCLUSIONS MBL-2 genotypes associated with high MBL serum levels represent a risk factor for NEC. This finding, together with the MBL expression in bowel tissue, supports a role for MBL in the pathogenesis of NEC.
Collapse
|
23
|
Rentea RM, Liedel JL, Welak SR, Cassidy LD, Mayer AN, Pritchard KA, Oldham KT, Gourlay DM. Intestinal alkaline phosphatase administration in newborns is protective of gut barrier function in a neonatal necrotizing enterocolitis rat model. J Pediatr Surg 2012; 47:1135-42. [PMID: 22703783 DOI: 10.1016/j.jpedsurg.2012.03.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 03/05/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Previously, we have shown that supplementation of intestinal alkaline phosphatase (IAP) decreased severity of necrotizing enterocolitis (NEC)-associated intestinal injury. We hypothesized that IAP administration is protective of intestinal epithelial barrier function in a dose-dependent manner. METHODS Control rat pups were vaginally delivered and breast-fed. Premature rats were divided into 4 groups: formula fed with lipopolysaccharide and hypoxia (NEC) or additional daily bovine IAP 40, 4, or 0.4 U/kg (NEC + IAP 40 U, IAP 4 U, or IAP 0.4 U). RESULTS Necrotizing enterocolitis is associated with decreased IAP protein expression and activity. Supplemental IAP increases IAP activity in intestinal homogenates and decreased NEC injury score in a dose-dependent manner. Intestinal injury as measured by fluorescein isothiocyanate-dextran flux from ileal loops showed increased permeability vs control, but supplemental IAP reversed this. Tight junction proteins claudin-1, claudin-3, occludin, and zonula occludin 1 were elevated in the NEC and IAP-treated groups with differences in expression patterns. No differences in messenger RNA levels were observed on postinjury day 3. Intestinal alkaline phosphatase administration decreases intestinal NEC injury in a dose-dependent manner. CONCLUSION Early enteral supplemental IAP may reduce NEC-related injury and may be useful for preserving the intestinal epithelial barrier function.
Collapse
|
24
|
Perng W, Cnattingius S, Iliadou A, Villamor E. Perinatal characteristics and risk of polio among Swedish twins. Paediatr Perinat Epidemiol 2012; 26:218-25. [PMID: 22471681 DOI: 10.1111/j.1365-3016.2012.01268.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Prenatal exposure to adverse environmental conditions is related to increased adult mortality in regions where infections are highly prevalent, yet there is little evidence of the impact of perinatal conditions on the risk of severe infections throughout life. Using prospectively collected data from 21 604 like-sexed Swedish twins of known zygosity born in 1926-1958, we examined the risk of polio in relation to perinatal characteristics using cohort and nested co-twin case-control analyses. Polio incidence was determined through an interview in 1998, and linkage with the Swedish national inpatient and death registries. There were 133 cases of polio. In the cohort analysis, birth length, birthweight and head circumference were positively associated with polio risk. After adjustment for sex, birth year, gestational age at birth and within-twin pair correlations, twins of shortest length (<44 cm) had a 67% ([95% CI: 6%, 88%]; P=0.04) lower risk of polio compared with the reference group (47-49 cm). After additional adjustment for birth length, every 100-g increase in birthweight was related to a 34% increased risk of polio ([95% CI: -1%, 82%]; P=0.06), and every 10-mm increase in head circumference was related to a 17% greater risk of polio ([95% CI: 5%, 31%]; P=0.004). In co-twin control analyses among 226 disease-discordant twins, birth length, birthweight and head circumference were 0.3 cm (P=0.19), 84 g (P=0.07) and 3 mm (P=0.08) higher in cases than controls, respectively. Similar associations were observed among monozygotic (n=84) and dizygotic (n=142) twins. These findings suggest that early intrauterine growth restriction may be inversely related to the incidence of polio.
Collapse
Affiliation(s)
- Wei Perng
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
25
|
Liu Y, Fatheree NY, Mangalat N, Rhoads JM. Lactobacillus reuteri strains reduce incidence and severity of experimental necrotizing enterocolitis via modulation of TLR4 and NF-κB signaling in the intestine. Am J Physiol Gastrointest Liver Physiol 2012; 302:G608-17. [PMID: 22207578 PMCID: PMC3311308 DOI: 10.1152/ajpgi.00266.2011] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis (NEC) is the leading gastrointestinal cause of mortality and morbidity in the premature infant. Premature infants have a delay in intestinal colonization by commensal bacteria and colonization with potentially pathogenic organisms. Lactobacillus reuteri is a probiotic that inhibits enteric infections, modulates the immune system, and may be beneficial to prevent NEC. In previous studies, L. reuteri strains DSM 17938 and ATCC PTA 4659 differentially modulated inflammation in vitro; however, the strains had equivalent anti-inflammatory responses in LPS feeding-induced ileitis in neonatal rats in vivo. The impact of these two strains in the prevention of NEC has not been previously investigated. NEC was induced in newborn rats by orogastric formula feeding and exposure to hypoxia. L. reuteri was added to the formula to prevent NEC. NEC score, Toll-like receptor (TLR)-signaling genes, phospho-IκB activity, and cytokine levels in the intestine were examined. Both strains significantly increased survival rate and decreased the incidence and severity of NEC, with optimal effects from DSM 17938. In response to probiotic, mRNA expression of IL-6, TNF-α, TLR4, and NF-κB was significantly downregulated, while mRNA levels of anti-inflammatory cytokine IL-10 were significantly upregulated. In parallel, L. reuteri treatment led to decrease intestinal protein levels of TLR4 and cytokine levels of TNF-α and IL-1β in newborn rats with NEC. Both strains significantly inhibited not only intestinal LPS-induced phospho-IκB activity in an ex vivo study but also decreased the levels of phospho-IκB in the intestines of NEC rat model. Cow milk formula feeding produced a similar but milder proinflammatory profile in the intestine that was also ameliorated by 17938. Our studies demonstrate that each of the two L. reuteri strains has potential therapeutic value in our NEC model and in enteritis associated with cow milk feeding. These results support the concept that L. reuteri may represent a valuable treatment to prevent NEC.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Nicole Y. Fatheree
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Nisha Mangalat
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Jon Marc Rhoads
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| |
Collapse
|
26
|
P-glycoprotein induction by breast milk attenuates intestinal inflammation in experimental necrotizing enterocolitis. J Transl Med 2011; 91:1668-79. [PMID: 21788941 PMCID: PMC3909679 DOI: 10.1038/labinvest.2011.113] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
P-glycoprotein (Pgp), a product of the multi-drug resistance gene MDR1a, is a broad specificity efflux ATP cassette transmembrane transporter that is predominantly expressed in epithelial tissues. Because mdr1a(-/-) mice tend to develop spontaneous colitis in bacteria-dependent manner, Pgp is believed to have a role in protection of the intestinal epithelium from luminal bacteria. Here we demonstrate that levels of Pgp in the small intestine of newborn rodents dramatically increase during breastfeeding, but not during formula feeding (FF). In rats and mice, levels of intestinal Pgp peak on days 3-7 and 1-5 of breastfeeding, respectively. The mdr1a(-/-) neonatal mice subjected to FF, hypoxia, and hypothermia have significantly higher incidence and pathology, as well as significantly earlier onset of necrotizing enterocolitis (NEC) than congenic wild type mice. Breast-fed mdr1a(-/-) neonatal mice are also more susceptible to intestinal damage caused by the opportunistic pathogen Cronobacter sakazakii that has been associated with hospital outbreaks of NEC. Breast milk, but not formula, induces Pgp expression in enterocyte cell lines in a dose- and time-dependent manner. High levels of ectopically expressed Pgp protect epithelial cells in vitro from apoptosis induced by C. sakazakii. Taken together, these results show that breast milk-induced expression of Pgp may have a role in the protection of the neonatal intestinal epithelium from injury associated with nascent bacterial colonization.
Collapse
|
27
|
Stockinger S, Hornef MW, Chassin C. Establishment of intestinal homeostasis during the neonatal period. Cell Mol Life Sci 2011; 68:3699-712. [PMID: 21952827 PMCID: PMC11114965 DOI: 10.1007/s00018-011-0831-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 09/07/2011] [Accepted: 09/07/2011] [Indexed: 12/15/2022]
Abstract
The intestinal mucosa faces the challenge of regulating the balance between immune tolerance towards commensal bacteria, environmental stimuli and food antigens on the one hand, and induction of efficient immune responses against invading pathogens on the other hand. This regulatory task is of critical importance to prevent inappropriate immune activation that may otherwise lead to chronic inflammation, tissue disruption and organ dysfunction. The most striking example for the efficacy of the adaptive nature of the intestinal mucosa is birth. Whereas the body surfaces are protected from environmental and microbial exposure during fetal life, bacterial colonization and contact with potent immunostimulatory substances start immediately after birth. In the present review, we summarize the current knowledge on the mechanisms underlying the transition of the intestinal mucosa during the neonatal period leading to the establishment of a stable, life-long host-microbial homeostasis. The environmental exposure and microbial colonization during the neonatal period, and also the influence of maternal milk on the immune protection of the mucosa and the role of antimicrobial peptides, are described. We further highlight the molecular mechanisms of innate immune tolerance in neonatal intestinal epithelium. Finally, we link the described immunoregulatory mechanisms to the increased susceptibility to inflammatory and infectious diseases during the neonatal period.
Collapse
Affiliation(s)
- Silvia Stockinger
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| | - Mathias W. Hornef
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| | - Cécilia Chassin
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
28
|
Gregory KE, Deforge CE, Natale KM, Phillips M, Van Marter LJ. Necrotizing enterocolitis in the premature infant: neonatal nursing assessment, disease pathogenesis, and clinical presentation. Adv Neonatal Care 2011; 11:155-64; quiz 165-6. [PMID: 21730907 PMCID: PMC3759524 DOI: 10.1097/anc.0b013e31821baaf4] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Necrotizing enterocolitis (NEC) remains one of the most catastrophic comorbidities associated with prematurity. In spite of extensive research, the disease remains unsolved. The aims of this article are to present the current state of the science on the pathogenesis of NEC, summarize the clinical presentation and severity staging of the disease, and highlight the nursing assessments required for early identification of NEC and ongoing care for infants diagnosed with this gastrointestinal disease. The distributions of systemic and intestinal clinical signs that are most sensitive to nursing assessment and associated with Bell Staging Criteria are presented. These descriptive data are representative of 117 cases of NEC diagnosed in low-gestational-age infants (<29 weeks' gestation). The data highlight the clinical signs most commonly observed in infants with NEC and thus provide NICU nurses an evidence-based guide for assessment and care of infants with NEC.
Collapse
MESH Headings
- Enteral Nutrition
- Enterocolitis, Necrotizing/complications
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/therapy
- Humans
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/therapy
- Nursing Assessment
- Risk Factors
- Severity of Illness Index
- Treatment Outcome
Collapse
Affiliation(s)
- Katherine E Gregory
- Boston College, William F. Connell School of Nursing, Chestnut Hill, Massachusetts 02467, USA.
| | | | | | | | | |
Collapse
|
29
|
Bucher BT, McDuffie LA, Shaikh N, Tarr PI, Warner BB, Hamvas A, White FV, Erwin CR, Warner BW. Bacterial DNA content in the intestinal wall from infants with necrotizing enterocolitis. J Pediatr Surg 2011; 46:1029-33. [PMID: 21683193 PMCID: PMC3118995 DOI: 10.1016/j.jpedsurg.2011.03.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Accepted: 03/26/2011] [Indexed: 11/17/2022]
Abstract
PURPOSE The objective of our study was to quantify mucosal bacterial DNA within specimens from neonates undergoing small bowel resection for necrotizing enterocolitis (NEC). METHODS We obtained clinical information and pathologic specimens from all infants diagnosed with NEC who underwent surgical treatment at our institution from 1999 to 2008. Bacterial and human DNA were isolated from paraffin-embedded surgical specimens, and real time polymerase chain reaction was used to amplify bacterial and human genes. Linear regression was used to quantify the amount of human and bacterial DNA in our specimens. RESULTS From a cohort of 50 infants, we identified 23 infants who underwent both surgical resection and subsequent intestinal reanastomosis. Thirteen (59%) of the neonates had Bell stage III NEC, and 9 (41%) had stage II. There was significantly more bacterial DNA in the resection specimens than in the reanastomosis specimens. This corresponds to a median (interquartile range) increase of 1.81 (1.11-4.69)-fold bacterial DNA in the resection specimen compared with the reanastomosis specimen (P < .05). CONCLUSION There is more bacterial DNA in infants with acute NEC compared with the same infants after the NEC had clinically resolved. These findings underscore the potential relevance of adherent or invasive bacteria across the bowel wall in the pathogenesis of NEC.
Collapse
Affiliation(s)
- Brian T. Bucher
- Division of Pediatric Surgery, Department of Surgery Washington University School of Medicine, St. Louis, MO
| | - Lucas A. McDuffie
- Division of Pediatric Surgery, Department of Surgery Washington University School of Medicine, St. Louis, MO
| | - Nurmohammad Shaikh
- Divison of Gastroenterology Washington University School of Medicine, St. Louis, MO
| | - Phillip I. Tarr
- Divison of Gastroenterology Washington University School of Medicine, St. Louis, MO
| | - Barbara B. Warner
- Newborn Medicine, Department of Pediatrics Washington University School of Medicine, St. Louis, MO
| | - Aaron Hamvas
- Newborn Medicine, Department of Pediatrics Washington University School of Medicine, St. Louis, MO
| | - Francis V. White
- Department of Pathology and Immunology; Washington University School of Medicine, St. Louis, MO
| | - Christopher R. Erwin
- Division of Pediatric Surgery, Department of Surgery Washington University School of Medicine, St. Louis, MO
| | - Brad W. Warner
- Division of Pediatric Surgery, Department of Surgery Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
30
|
The NFKB1 (g.-24519delATTG) variant is associated with necrotizing enterocolitis (NEC) in premature infants. J Surg Res 2011; 169:e51-7. [PMID: 21529841 DOI: 10.1016/j.jss.2011.03.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 02/11/2011] [Accepted: 03/04/2011] [Indexed: 01/04/2023]
Abstract
OBJECTIVE While it is known that gene-environment interactions contribute to necrotizing enterocolitis (NEC) pathogenesis, characterization of genetic risk-factors that can predict NEC in preterm infants remains nascent. We hypothesized that altered intestinal immune responses arising from sequence variation in the toll-like receptor (TLR) pathway genes contribute to NEC susceptibility. MATERIALS AND METHODS Very low birth weight (VLBW) infants were recruited prospectively in a multi-center, cohort study involving collection of blood samples along with collation of clinical information. DNA obtained from blood samples was used to genotype nine single nucleotide polymorphisms (SNPs) in eight TLR pathway genes by single-base extension. Prevalence of the variant allele was compared between cases and controls using Fisher's exact test. RESULTS In our cohort of 271 infants, 15 infants (5.6%) developed NEC, and five died from it. Infants with NEC were less mature (P < 0.001), and were more likely to be African-American (P = 0.007). SNPs in the TLR2, TLR4, TLR5, TLR9, IRAK1, and TIRAP genes were not associated with NEC. The NFKB1 (g.-24519delATTG) variant was present in all infants with NEC but only in 65% of infants without NEC (P = 0.003), while the NFKBIA (g.-1004A>G) variant was present in 13.3% of infants with NEC but in 49% of infants without NEC (P = 0.007). After correcting for multiple comparisons, the NFKB1 and NFKBIA variants remained associated with NEC (P < 0.05). CONCLUSIONS These data suggest that TLR genetic variants can alter susceptibility to NEC in VLBW infants and support the hypothesis that genetically programmed differences in the innate immune response contribute to NEC pathogenesis.
Collapse
|
31
|
Murgas Torrazza R, Suryawan A, Gazzaneo MC, Orellana RA, Frank JW, Nguyen HV, Fiorotto ML, El-Kadi S, Davis TA. Leucine supplementation of a low-protein meal increases skeletal muscle and visceral tissue protein synthesis in neonatal pigs by stimulating mTOR-dependent translation initiation. J Nutr 2010; 140:2145-52. [PMID: 20962152 PMCID: PMC2981001 DOI: 10.3945/jn.110.128421] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Protein synthesis and eukaryotic initiation factor (eIF) activation are increased in skeletal muscle of neonatal pigs parenterally infused with amino acids. Leucine appears to be the most effective single amino acid to trigger these effects. To examine the response to enteral leucine supplementation, overnight food-deprived 5-d-old pigs were gavage fed at 0 and 60 min a: 1) low-protein diet (LP); 2) LP supplemented with leucine (LP+L) to equal leucine in the high-protein diet (HP); or 3) HP diet. Diets were isocaloric and equal in lactose. Fractional protein synthesis rates and translation initiation control mechanisms were examined in skeletal muscles and visceral tissues 90 min after feeding. Protein synthesis rates in longissimus dorsi, gastrocnemius, and masseter muscles, heart, jejunum, kidney, and pancreas, but not liver, were greater in the LP+L group compared with the LP group and did not differ from the HP group. Feeding LP+L and HP diets compared with the LP diet increased phosphorylation of mammalian target of rapamycin (mTOR), 4E-binding protein 1, ribosomal protein S6 kinase-1, and eIF4G and formation of the active eIF4E·eIF4G complex in longissimus dorsi muscle. In all tissues except liver, activation of mTOR effectors increased in pigs fed LP+L and HP vs. LP diets. Our results suggest that leucine supplementation of a low-protein meal stimulates protein synthesis in muscle and most visceral tissues to a rate similar to that achieved by feeding a high-protein meal and this stimulation involves activation of mTOR downstream effectors.
Collapse
Affiliation(s)
- Roberto Murgas Torrazza
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Agus Suryawan
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Maria C. Gazzaneo
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Renán A. Orellana
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Jason W. Frank
- Department of Animal Science, University of Arkansas, Fayetteville, AR 72701
| | - Hanh V. Nguyen
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Marta L. Fiorotto
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Samer El-Kadi
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Teresa A. Davis
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
32
|
Role of postnatal acquisition of the intestinal microbiome in the early development of immune function. J Pediatr Gastroenterol Nutr 2010; 51:262-73. [PMID: 20639773 PMCID: PMC2932839 DOI: 10.1097/mpg.0b013e3181e1a114] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Therapy with broad-spectrum antibiotics is a common practice for premature infants. This treatment can reduce the biodiversity of the fecal microbiota and may be a factor in the cause of necrotizing enterocolitis. In contrast, probiotic treatment of premature infants reduces the incidence of necrotizing enterocolitis. We hypothesized that 1 mechanism for these observations is the influence of bacteria on postnatal development of the mucosal immune system. MATERIALS AND METHODS Expression of immune molecules and microbial sensors was investigated in the postnatal mouse gastrointestinal tract by real-time polymerase chain reaction. Subsequently, 2-week-old specific pathogen-free and microbial-reduced (MR; antibiotic treated) mice were compared for immune molecule and microbial sensor expression, mesenteric lymph node T-cell numbers and activation, intestinal barrier function/permeability, systemic lymphocyte numbers, and T-cell phenotype commitment. RESULTS Toll-like receptor 2, 4, and 5 expression was highest in 2-week-old specific pathogen-free mice, and this expression was decreased in MR mice. There was no difference in intestinal tight-junctional function, as evaluated by fluorescein isothiocyanate-dextran uptake, but MR mice had increased bacterial translocation across the intestinal epithelial barrier. MR mice had significantly fewer splenic B cells and mesenteric lymph node CD4+ T cells, but there were normal numbers of splenic T cells. These systemic T cells from MR mice produced more interleukin-4 and less interferon-gamma and IL-17, indicative of maintenance of the fetal, T-helper cell type 2 phenotype. CONCLUSIONS The present study shows that intestinal commensal microbiota have an influence on early postnatal immune development. Determining specific bacteria and/or bacterial ligands critical for this development could provide insight into the mechanisms by which broad-spectrum antibiotics and/or probiotic therapy influence the development of the mucosal immune system and mucosal-related diseases.
Collapse
|
33
|
Emami CN, Petrosyan M, Giuliani S, Williams M, Hunter C, Prasadarao NV, Ford HR. Role of the host defense system and intestinal microbial flora in the pathogenesis of necrotizing enterocolitis. Surg Infect (Larchmt) 2010; 10:407-17. [PMID: 19943775 DOI: 10.1089/sur.2009.054] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating disease that affects primarily the intestine of premature infants. Despite recent advances in neonatology, NEC remains a major cause of morbidity and mortality in neonates. Neonatal mucosal defenses and adherence of bacterial pathogens may play an important role in the pathogenesis of NEC. METHODS Review and synthesis of pertinent literature. RESULTS Putative factors that have been implicated in the pathogenesis of NEC include abnormal patterns of gut colonization by bacteria, immaturity of the host immune system and mucosal defense mechanisms, intestinal ischemia, formula feeding, and loss of intestinal epithelial barrier integrity. CONCLUSION Host defenses and intestinal microbial ecology are believed to play important roles in the pathogenesis of NEC. Commensal bacteria and probiotic therapy may be of therapeutic utility in the maintenance of the gut epithelial barrier.
Collapse
Affiliation(s)
- Claudia N Emami
- Department of Surgery, Childrens Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, California 90027, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Molecular modulation of intestinal epithelial barrier: contribution of microbiota. J Biomed Biotechnol 2010; 2010:305879. [PMID: 20150966 PMCID: PMC2817557 DOI: 10.1155/2010/305879] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 10/28/2009] [Indexed: 12/16/2022] Open
Abstract
The daunting task required of the gut-barrier to prevent luminal pathogens and harmful substances from entering into the internal milieu and yet promoting digestion and absorption of nutrients requires an exquisite degree of coordination between the different architectural units of this barrier. The complex integration and execution of these functions are superbly carried out by the intestinal mucosal (IM) surface. Exposed to trillions of luminal microbes, the IM averts threats by signaling to the innate immune system, through pattern recognition receptors (PRR), to respond to the commensal bacteria by developing tolerance (hyporesponsiveness) towards them. This system also acts by protecting against pathogens by elaborating and releasing protective peptides, cytokines, chemokines, and phagocytic cells. The IM is constantly sampling luminal contents and making molecular adjustments at its frontier. This article describes the topography of the IM and the mechanisms of molecular adjustments that protect the internal milieu, and also describes the role of the microbiota in achieving this goal.
Collapse
|
35
|
Wolfs TGAM, Derikx JPM, Hodin CMIM, Vanderlocht J, Driessen A, de Bruïne AP, Bevins CL, Lasitschka F, Gassler N, van Gemert WG, Buurman WA. Localization of the lipopolysaccharide recognition complex in the human healthy and inflamed premature and adult gut. Inflamm Bowel Dis 2010; 16:68-75. [PMID: 20014022 DOI: 10.1002/ibd.20995] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Microbiota in the intestinal lumen provide an abundant source of potentially detrimental antigens, including lipopolysaccharide (LPS), a potent immunostimulatory product of Gram-negative bacteria recognized by the host via TLR-4 and MD-2. An aberrant immune response to LPS or other bacterial antigens has been linked to inflammatory bowel disease (IBD) and necrotizing enterocolitis (NEC). METHODS We investigated which cells express MD-2 in the normal and inflamed ileum from neonates and adults by immunohistochemistry. Moreover, MD-2 and TLR4 mRNA expression in normal adult ileum was studied by reverse-transcription polymerase chain reaction (RT-PCR) on cells isolated by laser capture microdissection. RESULTS Premature infants did not show MD-2 expression either in epithelial cells or in the lamina propria. Similarly, MD-2 was absent in epithelial cells and lamina propria inflammatory cells in preterm infants with NEC. MD-2 protein in the healthy term neonatal and adult ileum was predominantly expressed by Paneth cells and some resident inflammatory cells in the lamina propria. MD-2 and TLR-4 mRNA expression was restricted to crypt cells. Also in IBD, Paneth cells were still the sole MD-2-expressing epithelial cells, whereas inflammatory cells (mainly plasma cells) were responsible for the vast majority of the MD-2 expression. CONCLUSIONS The absence of MD-2 in the immature neonatal gut suggests impaired LPS sensing, which could predispose neonates to NEC upon microbial colonization of the immature intestine. The apparent expression of MD-2 by Paneth cells supports the critical concept that these cells respond to luminal bacterial products in order to maintain homeostasis with the intestinal microbiota in vivo.
Collapse
Affiliation(s)
- Tim G A M Wolfs
- Department of Surgery, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Salzman NH, Bevins CL. Negative interactions with the microbiota: IBD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 635:67-78. [PMID: 18841704 DOI: 10.1007/978-0-387-09550-9_6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mucosal surfaces are colonized by a complex microbiota that provides beneficial functions under normal physiological conditions, but is capable of contributing to chronic inflammatory disease in susceptible individuals. Of the mucosal tissues, the mammalian intestine harbors an especially high number of microbes with a remarkable diversity. Inflammatory bowel disease (IBD) is a group of chronic relapsinginflammatory disorders of the intestinal mucosa. Evidence from human studies and animal models provides compelling support that intestinal microbes play a key role in disease pathogenesis. While the existence a specific causative pathogen is possible, it appears more likely that intestinal microbes normally present as commensal microbiota may trigger inflammation and perpetuate disease in genetically susceptible individuals. There may be also a shift in the makeup of the commensal flora to a nonphysiologic composition that is more prone to disease (termed dysbiosis). Evidence supports that genetic susceptibility stems from one or more defects in mucosal immune functions, including microbe recognition, barrier function, intercellular communication and antimicrobial effector mechanisms. It is quite plausible to imagine that the chronic inflammation of IBD may in some cases be a normal immune response to an abnormal adherent invasive microbiota and in other cases an over exuberant immune response to an otherwise normal commensal microbiota.
Collapse
Affiliation(s)
- Nita H Salzman
- Department of Pediatrics, Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
37
|
Schlapbach LJ, Aebi C, Fisch U, Ammann RA, Otth M, Bigler S, Nelle M, Berger S, Kessler U. Higher cord blood levels of mannose-binding lectin-associated serine protease-2 in infants with necrotising enterocolitis. Pediatr Res 2008; 64:562-6. [PMID: 18596574 DOI: 10.1203/pdr.0b013e3181841335] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Necrotising enterocolitis (NEC) causes significant morbidity and mortality in premature infants. The role of innate immunity in the pathogenesis of NEC remains unclear. Mannose-binding lectin (MBL) recognizes microorganisms and activates the complement system via MBL-associated serine protease-2 (MASP-2). The aim of this study was to investigate whether MBL and MASP-2 are associated with NEC. This observational case-control study included 32 infants with radiologically confirmed NEC and 64 controls. MBL and MASP-2 were measured in cord blood using ELISA. Multivariate logistic regression was performed. Of the 32 NEC cases (median gestational age, 30.5 wk), 13 (41%) were operated and 5 (16%) died. MASP-2 cord blood concentration ranged from undetectable (<10 ng/mL) to 277 ng/mL. Eighteen of 32 (56%) NEC cases had higher MASP-2 levels (> or =30 ng/mL) compared with 22 of 64 (34%) controls (univariate OR 2.46; 95% CI 1.03-5.85; p = 0.043). Higher cord blood MASP-2 levels were significantly associated with an increased risk of NEC in multivariate analysis (OR 3.00; 95% CI 1.17-7.93; p = 0.027). MBL levels were not associated with NEC (p = 0.64). In conclusion, infants later developing NEC had significantly higher MASP-2 cord blood levels compared with controls. Higher MASP-2 may favor complement-mediated inflammation and could thereby predispose to NEC.
Collapse
|
38
|
Schnabl KL, Aerde JEV, Thomson ABR, Clandinin MT. Necrotizing enterocolitis: A multifactorial disease with no cure. World J Gastroenterol 2008; 14:2142-61. [PMID: 18407587 PMCID: PMC2703838 DOI: 10.3748/wjg.14.2142] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Necrotizing enterocolitis is an inflammatory bowel disease of neonates with significant morbidity and mortality in preterm infants. Due to the multifactorial nature of the disease and limitations in disease models, early diagnosis remains challenging and the pathogenesis elusive. Although preterm birth, hypoxic-ischemic events, formula feeding, and abnormal bacteria colonization are established risk factors, the role of genetics and vasoactive/inflammatory mediators is unclear. Consequently, treatments do not target the specific underlying disease processes and are symptomatic and surgically invasive. Breast-feeding is the most effective preventative measure. Recent advances in the prevention of necrotizing enterocolitis have focused on bioactive nutrients and trophic factors in human milk. Development of new disease models including the aspect of prematurity that consistently predisposes neonates to the disease with multiple risk factors will improve our understanding of the pathogenesis and lead to discovery of innovative therapeutics.
Collapse
|
39
|
Cichocki M, Singer G, Beyerlein S, Zeder SL, Schober P, Höllwarth M. A case of necrotizing enterocolitis associated with adenovirus infection in a term infant with 22q11 deletion syndrome. J Pediatr Surg 2008; 43:e5-8. [PMID: 18405699 DOI: 10.1016/j.jpedsurg.2007.11.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 11/12/2007] [Accepted: 11/14/2007] [Indexed: 11/26/2022]
Abstract
Infections with adenoviruses are a common problem in the pediatric population. Normally asymptomatic to mild, those infections tend to take a more severe course in immunocompromised patients. 22q11 deletion syndrome (22q11DS) represents a common genetic disorder causing immunodeficiency from thymic hypoplasia or aplasia, heart defects, a characteristic facial appearance, and velopharyngeal dysfunction. Necrotizing enterocolitis (NEC) is a frequent gastrointestinal emergency observed in neonatal intensive care units. The occurrence of NEC is more prevalent in preterm infants. However, there are cases in term infants, but usually, they are associated with predisposing disorders. In this case report, a child is presented with 22q11DS that postnatally developed NEC associated with an adenoviral infection. Although other viruses such as toroviruses or cytomegaloviruses have been implicated in the pathogenesis of NEC in preterm infants, we could not find any report in the recent medical literature describing an association between adenoviral infections, NEC, and 22q11DS in a term infant.
Collapse
Affiliation(s)
- Martin Cichocki
- Department of Pediatric Surgery, Medical University of Graz, 8036 Graz, Austria.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
In necrotizing enterocolitis (NEC) the small (most often distal) and/or large bowel becomes injured, develops intramural air, and may progress to frank necrosis with perforation. Even with early, aggressive treatment, the progression of necrosis, which is highly characteristic of NEC, can lead to sepsis and death. This article reviews the current scientific knowledge related to the etiology and pathogenesis of NEC and discusses some possible preventive measures.
Collapse
|
41
|
Giannone PJ, Luce WA, Nankervis CA, Hoffman TM, Wold LE. Necrotizing enterocolitis in neonates with congenital heart disease. Life Sci 2008; 82:341-7. [DOI: 10.1016/j.lfs.2007.09.036] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 09/22/2007] [Accepted: 09/22/2007] [Indexed: 10/22/2022]
|
42
|
Turner D, Hammerman C, Rudensky B, Schlesinger Y, Wine E, Muise A, Schimmel MS. Low levels of procalcitonin during episodes of necrotizing enterocolitis. Dig Dis Sci 2007; 52:2972-6. [PMID: 17393328 DOI: 10.1007/s10620-007-9763-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Accepted: 01/03/2007] [Indexed: 12/13/2022]
Abstract
The pathogenesis of necrotizing enterocolitis (NEC) remains poorly understood. We aimed to assess the extent of bacterial infection in the pathogenesis of NEC using serial procalcitonin measurements. Blood samples were drawn during the first 4 days following every clinical event requiring a workup for presumed NEC. Eight episodes were confirmed as NEC, 7 of which showed procalcitonin levels <1 ng/ml at presentation and <1.3 ng/ml thereafter, comparable to 24 healthy controls. The one infant with elevated procalcitonin had bacteremia in addition to NEC. Procalcitonin levels of 24 matched septic infants were higher than those of NEC infants, peaking at 4.1 ng/ml. We conclude that low procalcitonin values are the rule during episodes of NEC and provide further evidence that overactive local immune response, and not active infection, is primarily responsible for the mucosal damage in NEC.
Collapse
Affiliation(s)
- Dan Turner
- Department of Pediatrics, Shaare Zedek Medical Center, Jerusalem, Israel.
| | | | | | | | | | | | | |
Collapse
|
43
|
Grave GD, Nelson SA, Walker WA, Moss RL, Dvorak B, Hamilton FA, Higgins R, Raju TNK. New therapies and preventive approaches for necrotizing enterocolitis: report of a research planning workshop. Pediatr Res 2007; 62:510-4. [PMID: 17667844 DOI: 10.1203/pdr.0b013e318142580a] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The National Institute of Child Health and Human Development and the Digestive Diseases Interagency Coordinating Committee held a workshop, chaired by Dr. W. Allan Walker, on July 10-11, 2006, to identify promising leads in necrotizing enterocolitis (NEC) research. The goals of the workshop were to identify new approaches to the prevention and treatment of NEC, to define basic and translational mechanisms of potential approaches to NEC, and to develop recommendations for clinical studies to reduce the incidence of NEC. Workshop participants implicated prematurity, introduction of enteral feedings, gastrointestinal bacterial colonization, gut motility, proinflammatory cytokines, impaired gut blood flow, and various neonatal complications in the pathogenesis of NEC. They concluded that a unifying hypothesis encompassing these pathogenetic factors is the uncontrolled exuberant inflammatory response to bacterial colonization that characterizes the intestine of premature infants. The inflammatory cascade appears to offer multiple targets for interventions with a variety of anti-inflammatory agents, including human milk and probiotics. Because of the rapidity with which the inflammatory response gets out of control in infants with NEC, workshop participants agreed that searching for ways to prevent NEC will be more rewarding than trying to identify ways to treat the condition once it has become established.
Collapse
Affiliation(s)
- Gilman D Grave
- Endocrinology, Nutrition and Growth Branch, National Institute of Child Health and Development/National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Anand RJ, Leaphart CL, Mollen KP, Hackam DJ. The role of the intestinal barrier in the pathogenesis of necrotizing enterocolitis. Shock 2007; 27:124-33. [PMID: 17224785 DOI: 10.1097/01.shk.0000239774.02904.65] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in neonates and is increasing in frequency because of recent advances in neonatal care. NEC develops in a stressed preterm infant in the setting of intestinal barrier disruption, systemic inflammation, and leads to, multisystem organ failure. The intestinal barrier lies at the interface between microbes within the intestinal lumen and the immune system of the host, and has both immunological and mechanical components. These components serve to protect the host from invading pathogens and, at the same time, provide a surface area for nutrient absorption. Factors that lead to impairments in the function of the intestinal barrier may predispose the host to the invasion of gut-derived microbes and to the development of systemic inflammatory disease. This process, termed "bacterial translocation," may be compounded during instances in which the mechanisms that regulate the repair of the intestinal barrier are disrupted. Bacterial translocation is of particular concern to the newborn patient, in which immaturity of the mechanical barrier and incomplete development of the host immune system combine to render the host at particular risk for the development of intestinal inflammation. This review will serve to provide an overview of recent evidence regarding the components of the intestinal barrier, and the mechanisms by which disruptions in barrier function may contribute to the pathogenesis of NEC.
Collapse
MESH Headings
- Animals
- Bacterial Translocation
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/microbiology
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/physiopathology
- Humans
- Immunity, Mucosal
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/immunology
- Infant, Premature, Diseases/microbiology
- Infant, Premature, Diseases/pathology
- Infant, Premature, Diseases/physiopathology
- Intestinal Absorption
- Intestines/immunology
- Intestines/microbiology
- Intestines/pathology
- Intestines/physiopathology
Collapse
Affiliation(s)
- Rahul J Anand
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
45
|
Salzman NH, Underwood MA, Bevins CL. Paneth cells, defensins, and the commensal microbiota: A hypothesis on intimate interplay at the intestinal mucosa. Semin Immunol 2007; 19:70-83. [PMID: 17485224 DOI: 10.1016/j.smim.2007.04.002] [Citation(s) in RCA: 263] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2007] [Accepted: 04/16/2007] [Indexed: 02/06/2023]
Abstract
Mucosal surfaces are colonized by a diverse and dynamic microbiota. Much investigation has focused on bacterial colonization of the intestine, home to the vast majority of this microbiota. Experimental evidence has highlighted that these colonizing microbes are essential to host development and homeostasis, but less is known about host factors that may regulate the composition of this ecosystem. While evidence shows that IgA has a role in shaping this microbiota, it is likely that effector molecules of the innate immune system are also involved. One hypothesis is that gene-encoded antimicrobial peptides, key elements of innate immunity throughout nature, have an essential role in this regulation. These effector molecules characteristically have activity against a broad spectrum of bacteria and other microbes. At mucosal surfaces, antimicrobial peptides may affect the numbers and/or composition of the colonizing microbiota. In humans and other mammals, defensins are a predominant class of antimicrobial peptides. In the small intestine, Paneth cells (specialized secretory epithelial cells) produce high quantities of defensins and several other antibiotic peptides and proteins. Data from murine models indicate that Paneth cell defensins play a pivotal role in defense from food and water-borne pathogens in the intestinal lumen. Recent studies in humans provide evidence that reduced Paneth cell defensin expression may be a key pathogenic factor in ileal Crohn's disease, a subgroup of inflammatory bowel disease (IBD), and changes in the colonizing microbiota may mediate this pathogenic mechanism. It is also possible that low levels of Paneth cell defensins, characteristic of normal intestinal development, may predispose premature neonates to necrotizing enterocolitis (NEC) through similar close links with the composition of the intestinal microbiota. Future studies to further define mechanisms by which defensins and other host factors regulate the composition of the intestinal microbiota will likely provide new insights into intestinal homeostasis and new therapeutic strategies for inflammatory and infectious diseases of the bowel.
Collapse
Affiliation(s)
- Nita H Salzman
- Department of Pediatrics, Division of Gastroenterology, The Medical College of Wisconsin, 8701 Watertown Plank Rd. Milwaukee, WI 53226, USA
| | | | | |
Collapse
|
46
|
Sharma R, Tepas JJ, Hudak ML, Mollitt DL, Wludyka PS, Teng RJ, Premachandra BR. Neonatal gut barrier and multiple organ failure: role of endotoxin and proinflammatory cytokines in sepsis and necrotizing enterocolitis. J Pediatr Surg 2007; 42:454-61. [PMID: 17336180 DOI: 10.1016/j.jpedsurg.2006.10.038] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND/PURPOSE Failure of the gut barrier and endotoxemia have been implicated in sepsis and multiple organ failure (MOF) syndromes in adults. The contributions of endotoxin (ETX) and proinflammatory cytokines (CKs) to the pathophysiology of disease and the outcomes of infants in the neonatal intensive care unit (NICU) are not clear. We measured ETX and CK concentrations in infants who presented with clinical signs of sepsis and/or necrotizing enterocolitis (NEC) to study their impact on MOF and outcomes. METHODS Blood samples from infants with signs of NEC and/or sepsis were collected for culture and determination of complete blood cell counts and concentrations of CKs (interleukin [IL]-1beta, tumor necrosis factor [TNF] alpha, and IL-6) and ETX at the onset of illness. Infants with signs of sepsis but without those of NEC were classified by blood culture results into a confirmed sepsis group (ie, positive culture) or a control group (ie, negative culture). Endotoxin concentrations were determined by chromogenic Limulus amebocyte lysate assay, and CK levels were quantitated by enzyme-linked immunoassay. Data are expressed as mean +/- SD and as odds ratios (ORs) with 95% confidence intervals (CIs). P values lower than .05 were considered to be significant. RESULTS There was no demographic or clinical difference among the NEC (n = 27), sepsis (n = 44), and control (n = 56) groups, except that fewer (P = .02) infants in the NEC group (11%) had received maternal milk feedings as compared with infants in the sepsis group (23%) and those in the control group (39%). Endotoxin concentrations were higher (P < .0001) in the NEC group (3.30 +/- 2.11) as compared with the sepsis group (0.67 +/- .86) and the control group (0.09 +/- 0.24). Generalized linear regression analysis using formula feeding, mechanical ventilation, and gram-negative bacteremia as covariates demonstrated that NEC increased ETX concentrations independently (r = .80; P < .0001). Endotoxemia correlated with higher concentrations of all 3 CKs (P < .0001). There was an inverse association between ETX and both platelet count (r = -0.30; P = .0003) and absolute neutrophil count (r = -0.29; P = .0009). Infants who died of MOF had higher concentrations of ETX (2.83 +/- 3.04 vs 0.67 +/- 1.04 EU/mL; P < .0001), IL-1beta (509 +/- 493 vs 106 +/- 223 pg/mL; P < .0001), IL-6 (416 +/- 308 vs 99 +/- 165 pg/mL; P < .0001), and TNF-alpha (503 +/- 449 vs 126 +/- 237 pg/mL; P < .0001) as compared with those without MOF. Eighty-six percent of the infants with MOF died. Multivariate logistic regression analysis demonstrated that higher ETX concentrations (OR = 2.47; 95% CI = 1.39-4.40; P = .002) and lower gestational age (OR = 1.41; 95% CI = 1.12-1.77; P = .003) predicted mortality. CONCLUSIONS Neonatal endotoxemia and release of proinflammatory CKs are important contributors to MOF and mortality in the NICU. Endotoxemia was most severe at the onset of illness among the infants with NEC, suggesting that gut barrier failure plays an important role in adverse outcomes in the NICU.
Collapse
Affiliation(s)
- Renu Sharma
- Department of Pediatrics, University of Florida at Jacksonville, Jacksonville, FL 32209, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Our understanding of the relationship between microbes and their animal hosts have changed dramatically in the last decade. The development of powerful new molecular methods as well as different animal models, particularly germ free rodents, has enabled precise characterisation of the ways in which commensal bacteria in the gastrointestinal tract and other areas interact with their hosts. It is now clear that animal development, far from being genetically determined is intimately bound up with the microbial flora of that particular animal. In germ free mice, the addition of Bacteroides thetaiotaomicron mediates the maturation and function of the gastrointestinal tract in several different ways. Similarly Bacteroides fragilis directs the development of the immune system both in and outside the gastrointestinal tract. The relevance of these findings and others to our understanding of human development and disease is discussed.
Collapse
Affiliation(s)
- Mark Wilks
- Department of Microbiology, 3rd Floor, Path and Pharm Block, 80 Newark St, E1 2ES, London, United Kingdom.
| |
Collapse
|
48
|
Abstract
Necrotizing enterocolitis is the most common gastrointestinal emergency of the neonate, affecting 5-10% of infants, yet the pathogenesis remains unclear. Widely accepted risk factors include prematurity, enteral feeds, bacterial colonization and mucosal injury. How these or other yet identified factors come together to create the classic clinical and pathologic features is the subject of much research. The activation of the cytokine cascade, in part by bacterial ligands, appears to play a key role in mucosal injury. Two mediators that may also contribute are platelet activating factor and intestinal toll-like receptors. Short chain fatty acids, the products of bacterial fermentation of carbohydrates, have been thought to cause mucosal injury. Overgrowth of pathogenic bacteria in the face of a decreased commensal population may play a key role. A current focus of clinical research involves probiotics, enterally fed forms of commsenal bacteria. This may set the stage for a healthier intestinal ecosystem and possibly, decreased risk of NEC.
Collapse
MESH Headings
- Combined Modality Therapy
- Enteral Nutrition
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/mortality
- Enterocolitis, Necrotizing/therapy
- Fatty Acids, Volatile/metabolism
- Female
- Fluid Therapy/methods
- Humans
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/mortality
- Infant, Premature, Diseases/therapy
- Inflammation Mediators/analysis
- Intestinal Mucosa/pathology
- Male
- Probiotics/administration & dosage
- Prognosis
- Risk Factors
- Severity of Illness Index
- Survival Rate
- Toll-Like Receptors/metabolism
Collapse
Affiliation(s)
- Kathleen Gibbs
- Division of Newborn Medicine, Department of Pediatrics, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
49
|
Abstract
Epithelial cell functions ultimately define the ability of the extremely low birth weight human fetus to survive outside of the uterus. These specialized epithelial cell capacities manage all human interactions with the ex utero world including: (i) lung mechanics, surface chemistry and gas exchange, (ii) renal tubular balance of fluid and electrolytes, (iii) barrier functions of the intestine and skin for keeping bacteria out and water in, plus enabling intestinal digestion, as well as (iv) maintaining an intact neuroepithelium lining of the ventricles of the brain and retina. In Part I of this two part review, the authors describe why the gut barrier is a clinically relevant model system for studying the complex interplay between innate and adaptive immunity, dendritic &epithelial cell interactions, intraepithelial lymphocytes, M-cells, as well as the gut associated lymphoid tissues where colonization after birth, clinician feeding practices, use of antibiotics as well as exposure to prebiotics, probiotics and maternal vaginal flora all program the neonate for a life-time of immune competence distinguishing "self" from foreign antigens. These barrier defense capacities become destructive during disease processes like necrotizing enterocolitis (NEC) when an otherwise maturationally normal, yet dysregulated and immature, immune defense system is associated with high levels of certain inflammatory mediators like TNFa. In Part II the authors discuss the rationale for why rhG-CSF has theoretical advantages in managing NEC or sepsis by augmenting neonatal neutrophil number, neutrophil expression of Fcg and complement receptors, as well as phagocytic function and oxidative burst. rhG-CSF also has potent anti-TNFa functions that may serve to limit extension of tissue destruction while not impairing bacterial killing capacity. Healthy, non-infected neutropenic and septic neonates differ in their ability to respond to rhG-CSF; however, no neonatal clinical trials to date have identified a clear clinical benefit of rhG-CSF therapy. This manuscript will review the literature and evidence available for identifying the ideal subject for cytokine treatment using NEC as the model disease target.
Collapse
Affiliation(s)
- Aryeh Simmonds
- Division of Newborn Medicine, The Regional Neonatal Center, Maria Fareri Children's Hospital of Westchester Medical Center, New York Medical College, Valhalla, NY 10595, USA
| | | |
Collapse
|
50
|
Abstract
Epithelial cell functions ultimately define the ability of the extremely low birth weight human fetus to survive outside of the uterus. These specialized epithelial cell capacities manage all human interactions with the ex utero world including: (i) lung mechanics, surface chemistry and gas exchange, (ii) renal tubular balance of fluid and electrolytes, (iii) barrier functions of the intestine and skin for keeping bacteria out and water in, plus enabling intestinal digestion, as well as (iv) maintaining an intact neuroepithelium lining of the ventricles of the brain and retina. In Part I of this two part review, the authors describe why the gut barrier is a clinically relevant model system for studying the complex interplay between innate and adaptive immunity, dendritic &epithelial cell interactions, intraepithelial lymphocytes, M-cells, as well as the gut associated lymphoid tissues where colonization after birth, clinician feeding practices, use of antibiotics as well as exposure to prebiotics, probiotics and maternal vaginal flora all program the neonate for a life-time of immune competence distinguishing "self" from foreign antigens. These barrier defense capacities become destructive during disease processes like necrotizing enterocolitis (NEC) when an otherwise maturationally normal, yet dysregulated and immature, immune defense system is associated with high levels of certain inflammatory mediators like TNFa. In Part II, the authors will discuss the theoretical advantages of using rhG-CSF in managing NEC or sepsis by augmenting neonatal neutrophil number and killing capacity including an unexpected, paradoxical and potent anti-TNFa function that may serve to limit extension of tissue destruction without impairing bacterial killing capacity. The authors conclude by arguing that NEC may be the ideal disease process for testing whether a clearly defined clinical benefit of cytokine therapy can prove beneficial.
Collapse
Affiliation(s)
- Aryeh Simmonds
- Division of Newborn Medicine, The Regional Neonatal Center, Maria Fareri Children's Hospital of Westchester Medical Center, New York Medical College, Valhalla, NY 10595, USA
| | | |
Collapse
|