1
|
Peng N, Fu L, Liang X, Lu Q. Risk factors of brain abscess in neonatal meningitis: a propensity score-matched study. Eur J Pediatr 2023; 182:2215-2223. [PMID: 36867235 DOI: 10.1007/s00431-023-04860-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 03/04/2023]
Abstract
Brain abscess is a rare but life-threatening complication of meningitis. The purpose of this study was to identify clinical features and potentially relevant factors of brain abscess in neonates with meningitis. This study was a propensity score-matched case-control study of neonates with brain abscess and meningitis in a tertiary pediatric hospital between January 2010 and December 2020. A total of 16 neonates with brain abscess were matched to 64 patients with meningitis. Demography, clinical characteristics, laboratory results, and pathogens were collected. Conditional logistic regression analyses were performed to identify the independent risk factors associated with brain abscess. The most common pathogen we found in the brain abscess group was Escherichia coli. Risk factors of brain abscess were identified: multidrug-resistant bacterial infection (OR, 11.204; 95% CI, 2.315-54.234; p = 0.003), C-reactive protein (CRP) > 50 mg/L (OR, 11.652; 95% CI, 1.799-75.470; p = 0.010). Conclusion: The risk factors of brain abscess are multidrug-resistant bacterial infection and CRP > 50 mg/L. Monitoring the level of CRP is essential. Bacteriological culture and rational use of antibiotics are necessary for the prevention of MDR bacterial infection as well as the occurrence of brain abscess. What is Known: • Morbidity and mortality of neonatal meningitis have declined, but brain abscess associated with neonatal meningitis is still life-threatening. What is New: • This study investigated relevant factors related to brain abscess. • It is important for neonatologists to perform prevention, early identification, and appropriate interventions for neonates with meningitis.
Collapse
Affiliation(s)
- Nan Peng
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Medical University, Chongqing, China
| | - Lizhen Fu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Medical University, Chongqing, China
| | - Xiaohua Liang
- Department of Clinical Epidemiology and Bioinformatics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Medical University, Chongqing, China
| | - Qi Lu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Provitera L, Tomaselli A, Raffaeli G, Crippa S, Arribas C, Amodeo I, Gulden S, Amelio GS, Cortesi V, Manzoni F, Cervellini G, Cerasani J, Menis C, Pesenti N, Tripodi M, Santi L, Maggioni M, Lonati C, Oldoni S, Algieri F, Garrido F, Bernardo ME, Mosca F, Cavallaro G. Human Bone Marrow-Derived Mesenchymal Stromal Cells Reduce the Severity of Experimental Necrotizing Enterocolitis in a Concentration-Dependent Manner. Cells 2023; 12:cells12050760. [PMID: 36899900 PMCID: PMC10000931 DOI: 10.3390/cells12050760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/10/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating gut disease in preterm neonates. In NEC animal models, mesenchymal stromal cells (MSCs) administration has reduced the incidence and severity of NEC. We developed and characterized a novel mouse model of NEC to evaluate the effect of human bone marrow-derived MSCs (hBM-MSCs) in tissue regeneration and epithelial gut repair. NEC was induced in C57BL/6 mouse pups at postnatal days (PND) 3-6 by (A) gavage feeding term infant formula, (B) hypoxia/hypothermia, and (C) lipopolysaccharide. Intraperitoneal injections of PBS or two hBM-MSCs doses (0.5 × 106 or 1 × 106) were given on PND2. At PND 6, we harvested intestine samples from all groups. The NEC group showed an incidence of NEC of 50% compared with controls (p < 0.001). Severity of bowel damage was reduced by hBM-MSCs compared to the PBS-treated NEC group in a concentration-dependent manner, with hBM-MSCs (1 × 106) inducing a NEC incidence reduction of up to 0% (p < 0.001). We showed that hBM-MSCs enhanced intestinal cell survival, preserving intestinal barrier integrity and decreasing mucosal inflammation and apoptosis. In conclusion, we established a novel NEC animal model and demonstrated that hBM-MSCs administration reduced the NEC incidence and severity in a concentration-dependent manner, enhancing intestinal barrier integrity.
Collapse
Affiliation(s)
- Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
- Correspondence: (G.R.); (G.C.); Tel.: +39-(02)-55032234 (G.C.); Fax: +39-(02)-55032217 (G.R. & G.C.)
| | - Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cristina Arribas
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Silvia Gulden
- Neonatal Intensive Care Unit, Sant’Anna Hospital, 22042 Como, Italy
| | - Giacomo Simeone Amelio
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Valeria Cortesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Francesca Manzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Gaia Cervellini
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Jacopo Cerasani
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Camilla Menis
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Nicola Pesenti
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, University of Milano-Bicocca, 20126 Milan, Italy
- Revelo Datalabs S.R.L., 20142 Milan, Italy
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco Maggioni
- Department of Pathology, Fondazione Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Caterina Lonati
- Center for Preclinical Investigation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Samanta Oldoni
- Center for Preclinical Investigation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Algieri
- Research and Development Unit, Postbiotica S.R.L., 20123 Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Pediatric Immunohematology Unit, BMT Program, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Maternal and Child Department, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence: (G.R.); (G.C.); Tel.: +39-(02)-55032234 (G.C.); Fax: +39-(02)-55032217 (G.R. & G.C.)
| |
Collapse
|
3
|
Kumar T, Dutta RR, Velagala VR, Ghosh B, Mudey A. Analyzing the Complicated Connection Between Intestinal Microbiota and Cardiovascular Diseases. Cureus 2022; 14:e28165. [PMID: 36148181 PMCID: PMC9482761 DOI: 10.7759/cureus.28165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/19/2022] [Indexed: 12/12/2022] Open
Abstract
Relentless human curiosity to understand the basis of every aspect of medical science has led humanity to unlock the deepest secrets about the physiology of human existence and, in the process, has reached milestones that a century ago could only be imagined. Recent ground-breaking breakthroughs have helped scientists and physicians all over the world to update the scientific basis of diseases and hence further improve treatment outcomes. According to recent studies, scientists have found a link between intestinal flora and the pathogenesis of diseases, including cardiovascular diseases. Any change in the typical habitat of gut microbiota has been shown to result in the culmination of various metabolic and cardiac diseases. Therefore, gut microbiota can be credited for influencing the course of the development of a disease. Any change in the composition and function of bacterial species living in the gut can result in both beneficial and harmful effects on the body. Gut microbiota achieves this role by numerous mechanisms. Generations of various metabolites like TMAO (trimethylamine N-oxide), increased receptibility of various bacterial antigens, and disruption of the enzyme action in various metabolic pathways like the bile acids pathway may result in the development of metabolic as well as cardiovascular diseases. Even if they may not be the only etiological factor in the pathogenesis of a disease, they may very well serve as a contributing factor in worsening the outcome of the condition. Studies have shown that they actively play a role in the progression of cardiovascular diseases like atherosclerotic plaque formation and rising blood pressure. The focus of this review article is to establish a relation between various cardiovascular diseases and gut microbiota. This could prove beneficial for clinicians, health care providers, and scientists to develop novel therapeutic algorithms while treating cardiac patients.
Collapse
|
4
|
Evaluation of Enterococcus faecalis, Lactobacillus acidophilus, and Lactobacillus plantarum in Biopsy Samples of Colorectal Cancer and Polyp Patients Compared to Healthy People. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2022. [DOI: 10.5812/archcid-116165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Colorectal cancer (CRC) is one of the leading causes of death in both men and women worldwide. According to different studies, infectious agents or microbiota dysbiosis can play a role in CRC progression. Objective: This study aimed to evaluate the prevalence of Enterococcus faecalis, Lactobacillus acidophilus, and Lactobacillus plantarum in people with polyps or CRC compared to healthy individuals. Methods: In this study, 60 biopsy samples were collected from three groups, including patients with CRC, polyps, and healthy people. The genomic DNA was extracted from the collected samples and amplified by polymerase chain reaction (PCR) to detect E. faecalis, L. acidophilus, and L. plantarum. In the next step, quantitative Real-Time PCR was used to evaluate the copy number of the bacteria in the studied groups. Results: There was no statistically significant difference between the studied groups regarding age and gender (P > 0.05). The mean number of E. faecalis was higher in patients with CRC than in patients with polyps and healthy individuals (P < 0.05). Also, the mean numbers of L. acidophilus and L. plantarum were higher in healthy individuals than in patients with polyps and CRC (P < 0.05). Conclusions: Our findings indicate that L. acidophilus and L. plantarum in people with a family history of CRC and patients with polyps may effectively prevent or reduce CRC progression.
Collapse
|
5
|
The effects of IGF-1 and erythropoietin on apoptosis and telomerase activity in necrotizing enterocolitis model. Pediatr Res 2021; 90:559-564. [PMID: 33096541 DOI: 10.1038/s41390-020-01195-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Apoptosis that occurs after hypoxia/reoxygenation (H/R) has an important role in the pathogenesis of necrotizing enterocolitis (NEC). Telomerase activity, showing the regeneration capacity, may also be important in the recovery process. Therefore, we aimed to investigate the effects of insulin-like growth factor-1 (IGF-1) and erythropoietin (EPO) on apoptosis and telomerase activity in an H/R model. METHODS Young mice were divided into four groups each containing ten Balb/c mice. Group 1 (H/R) were exposed to H/R; group 2 and group 3 were pretreated with IGF-1 and EPO, respectively, for 7 days before H/R. Group 4 served as control. Intestinal injury was evaluated by histological scoring and assessment of apoptosis was performed by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) test. Proapoptotic and antiapoptotic gene expressions and telomerase activity were analyzed by real-time PCR. RESULTS IGF-1- and EPO-treated animals had decreased histological damage and apoptosis, confirmed by TUNEL test and caspase activity. Telomerase activity was increased in these animals in addition to increased expression of antiapoptotic genes. However, proapoptotic gene expressions were not statistically different. CONCLUSIONS The protective effects of IGF-1 and EPO in H/R damage may be through increased expression of antiapoptotic genes and increased telomerase activity, especially for IGF-1. IMPACT This is a comprehensive study measuring various variables, namely IGF-1, EPO, apoptosis, apoptotic and antiapoptotic genes, and telomerase activity in the NEC model. The intestinal protective effects of IGF-1 and EPO in H/R damage may occur through increased expression of antiapoptotic genes and increased telomerase activity. To the best of our knowledge, telomerase activity has not been investigated in the NEC model before. Regarding our results, novel strategies may be implemented for the early definitive diagnosis, robust preventive measures, and effective treatment modalities for NEC.
Collapse
|
6
|
Zhu H, Lin Y, Liu Y. miR‑34a increases inflammation and oxidative stress levels in patients with necrotizing enterocolitis by downregulating SIRT1 expression. Mol Med Rep 2021; 24:664. [PMID: 34296298 DOI: 10.3892/mmr.2021.12303] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/24/2021] [Indexed: 11/06/2022] Open
Abstract
The miR‑34a/SIRT1 signaling axis is an important signaling axis in tumors and diseases. Notably, low SIRT1 expression in the intestinal tissues of patients with necrotizing enterocolitis (NEC) has been reported. However, whether miR‑34a/SIRT1 signaling as a target to protect the intestines during the NEC process is unclear and remains to be elucidated. Blood samples were collected from 30 patients with NEC, and an NEC rat model was used. The miR‑34a and SIRT1 gene and protein expression levels were assayed by qPCR and Western blotting method. The inflammatory cytokine levels and oxidative stress levels were detected using the ELISA method. The results demonstrated that birth weight, albumin and glucose concentrations were significantly decreased in the NEC patient group compared with the control group, but the C‑reactive protein (CRP) and procalcitonin (PCT) concentrations were significantly increased. The miR‑34a expression level was notably increased in the NEC group, but the SIRT1 expression level was markedly decreased. Notably, the miR‑34a was significantly correlated with NEC severity and the concentrations of CRP, PCT, IL‑6, TNF‑α, IL‑1β, IL‑8, MCP‑1, VCAM1 and malondialdehyde (MDA), but was significantly negatively correlated with SIRT1 gene expression and the concentration of IL‑10. Intestinal villi damage in NEC rats was decreased with miR‑34a inhibition and SIRT1 activation treatment by decreasing the levels of inflammatory cytokines, including IL‑6, TNF‑α, IL‑1β and IL‑8, and oxidative stress proteins, including MCP‑1, VCAM1, and MDA, as well as increasing the level of the anti‑inflammatory cytokine IL‑10. In addition, the results indicated that miR‑34a inhibition and SIRT1 activation strongly protected the intestine and decreased the damage caused by NEC, not only by decreasing the protein levels of SIRT1, TNF‑α, IL‑1β, IL‑6 and IL‑8, but also by increasing the IL‑10 protein levels. The miR‑34a inhibition and SIRT1 activation may decrease the damage caused by NEC by decreasing proinflammatory cytokines and oxidative stress proteins and by increasing the anti‑inflammatory cytokine pathway. Based on the aforementioned analysis, the miR‑34a and SIRT1 proteins may be potential novel therapeutic targets in NEC.
Collapse
Affiliation(s)
- Hui Zhu
- Department of NICU, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yan Lin
- Department of NICU, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yongle Liu
- Department of NICU, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
7
|
Gong C, Yang L, Liu K, Shen S, Zhang Q, Li H, Cheng Y. Effects of Antibiotic Treatment and Probiotics on the Gut Microbiome of 40 Infants Delivered Before Term by Cesarean Section Analysed by Using 16S rRNA Quantitative Polymerase Chain Reaction Sequencing. Med Sci Monit 2021; 27:e928467. [PMID: 33542172 PMCID: PMC7871509 DOI: 10.12659/msm.928467] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/19/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND This study aimed to investigate the effects on the gut microbiome of 40 infants delivered before term by cesarean section between antibiotic treatment and probiotics as assessed by 16S rRNA quantitative polymerase chain reaction (qPCR) sequencing. MATERIAL AND METHODS We divided 40 premature infants delivered by cesarean section into 4 groups according to exposure to antibiotics or probiotics: N group (No-probiotics and No-antibiotics), A group (antibiotics), P group (probiotics), and the AP group (antibiotics+probiotics). Fecal samples were collected on days 1, 3, and 10, and the microflora data were generated using 16S rRNA qPCR sequencing technology. The BugBase tool was used for phenotype prediction, the Tax4Fun tool was used for function prediction, and iPath software was used to predict the metabolic pathways of intestinal bacteria. RESULTS Antibiotics increased the abundance of pathogenic bacteria and reduced the replication and repair function (P=0.049), nucleotide metabolism function (P=0.047), and the purine metabolism pathways (P<0.05) of the gut microbiota. Probiotics increased the abundance of beneficial bacteria and the cellular community prokaryote function (P=0.042) and contributed to the Bifidobacteria biofilm formation. Probiotics alleviated the damage of antibiotics to the composition and function of the gut microbiota. CONCLUSIONS The findings from this study showed that antibiotic treatment of preterm infants born by cesarean section changed the gut microbiome, but that the use of probiotics could restore the normal microbiome, which supports that restoration of the normal gut microbiota may be achieved with probiotics.
Collapse
Affiliation(s)
- Chen Gong
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Kangkang Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Shichun Shen
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Qixing Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Han Li
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
8
|
Profile analysis reveals endogenous RNAs regulate necrotizing enterocolitis progression. Biomed Pharmacother 2020; 125:109975. [PMID: 32036223 DOI: 10.1016/j.biopha.2020.109975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 12/31/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is one of the most common and devastating gastrointestinal diseases in preterm newborns, and its underlying mechanisms remain unclear. Non-coding RNAs (ncRNAs) play critical roles in intestinal diseases; however, little is known about their roles in the development of NEC. To gain a deeper understanding of the pathophysiological mechanism of NEC, long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and mRNAs were detected in an NEC rat model. In total, 1820 lncRNAs, 118 miRNAs and 929 mRNAs were differentially expressed in NEC group. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that these molecules were enriched in apoptosis, autophagic cell death, TLR4 signaling pathway, Notch signaling pathway, and mTOR signaling pathway. These pathways are thought to be closely associated with NEC. Furthermore, a lncRNA-miRNA interaction network was constructed, and four of the novel, differentially expressed lncRNAs with large changes were randomly verified using quantitative polymerase chain reaction (qPCR). The GO and KEGG pathway analysis of these four lncRNAs showed that they were associated with the negative regulation of TLR4 signaling pathway and Notch signaling pathway. In conclusion, our study revealed that these differentially expressed lncRNAs may participate in the development of NEC via interactions with miRNAs and may serve as possible biomarkers and target genes in NEC.
Collapse
|
9
|
Wertheimer F, Arcinue R, Niklas V. Necrotizing Enterocolitis: Enhancing Awareness for the General Practitioner. Pediatr Rev 2019; 40:517-527. [PMID: 31575803 DOI: 10.1542/pir.2017-0338] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Necrotizing enterocolitis (NEC) has been recognized for well over 5 decades yet remains the most common life-threatening surgical emergency in the newborn. The incidence of NEC has decreased steadily in preterm and very-low-birthweight infants over several decades and is typically uncommon in term newborns and infants with a birthweight greater than 2,500 g. Evidence accumulating during the past decade, however, suggests that practitioners should consider NEC in this broader subset of term infants with chromosomal and congenital anomalies complicated by heart or gastrointestinal defects when signs and symptoms of feeding intolerance, abdominal illness, or sepsis are present. The short- and long-term consequences of NEC are devastating in all infants, and although early disease recognition and treatment are essential, promoting human milk feeding as a primary modality in prevention is critical. This article highlights our current understanding of the pathophysiology, the clinical presentation, the risk factors for NEC in term infants compared with premature infants, and the treatment of NEC and discusses strategies in the prevention of NEC. Finally, we review the long-term consequences of NEC and the importance of primary care practitioners in the long-term care of infants after hospitalization for NEC.
Collapse
Affiliation(s)
- Fiona Wertheimer
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, Los Angeles, CA.,Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Roxanne Arcinue
- Keck School of Medicine, University of Southern California, Los Angeles, CA.,Fetal and Neonatal Institute, Division of Neonatology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA
| | - Victoria Niklas
- Prolacta Bioscience Inc, Duarte, CA, and Department of Pediatrics, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
10
|
Zhuang L, Chen H, Zhang S, Zhuang J, Li Q, Feng Z. Intestinal Microbiota in Early Life and Its Implications on Childhood Health. GENOMICS, PROTEOMICS & BIOINFORMATICS 2019; 17:13-25. [PMID: 30986482 PMCID: PMC6522475 DOI: 10.1016/j.gpb.2018.10.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/07/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023]
Abstract
Trillions of microbes reside in the human body and participate in multiple physiological and pathophysiological processes that affect host health throughout the life cycle. The microbiome is hallmarked by distinctive compositional and functional features across different life periods. Accumulating evidence has shown that microbes residing in the human body may play fundamental roles in infant development and the maturation of the immune system. Gut microbes are thought to be essential for the facilitation of infantile and childhood development and immunity by assisting in breaking down food substances to liberate nutrients, protecting against pathogens, stimulating or modulating the immune system, and exerting control over the hypothalamic-pituitary-adrenal axis. This review aims to summarize the current understanding of the colonization and development of the gut microbiota in early life, highlighting the recent findings regarding the role of intestinal microbes in pediatric diseases. Furthermore, we also discuss the microbiota-mediated therapeutics that can reconfigure bacterial communities to treat dysbiosis.
Collapse
Affiliation(s)
- Lu Zhuang
- Affiliated Bayi Children's Hospital, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China; National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing 100700, China; Beijing Key Laboratory of Pediatric Organ Failure, Beijing 100700, China
| | - Haihua Chen
- Affiliated Bayi Children's Hospital, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China; The First Clinical Academy of Dalian Medical University, Dalian 116011, China
| | - Sheng Zhang
- Affiliated Bayi Children's Hospital, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China; National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing 100700, China; Beijing Key Laboratory of Pediatric Organ Failure, Beijing 100700, China
| | - Jiahui Zhuang
- College of the Environment, Northeast Normal University, Changchun 130117, China
| | - Qiuping Li
- Affiliated Bayi Children's Hospital, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China; National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing 100700, China; Beijing Key Laboratory of Pediatric Organ Failure, Beijing 100700, China.
| | - Zhichun Feng
- Affiliated Bayi Children's Hospital, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China; National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing 100700, China; Beijing Key Laboratory of Pediatric Organ Failure, Beijing 100700, China.
| |
Collapse
|
11
|
Yu R, Jiang S, Tao Y, Li P, Yin J, Zhou Q. Inhibition of HMGB1 improves necrotizing enterocolitis by inhibiting NLRP3 via TLR4 and NF-κB signaling pathways. J Cell Physiol 2019; 234:13431-13438. [PMID: 30618088 DOI: 10.1002/jcp.28022] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/30/2018] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To explore the relationship between high-mobility group box 1 (HMGB1) and NLR pyrin domain containing 3 (NLRP3) in the development of necrotizing enterocolitis (NEC). METHODS NEC rat models were constructed and treated with HMGB1 inhibitor glycyrrhizin (GL) with different concentration. An inflammatory condition of intestinal tissue in newborn NEC rats was observed by hematoxylin and eosin staining. The messenger RNA (mRNA) and protein expression of HMGB1, NLRP3, toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), and caspase 1 were determined by real-time polymerase chain reaction and western blot analysis, respectively. The content of interleukin (IL)-1β and tumor necrosis factor-α (TNF-α) was determined by enzyme-linked immunosorbent assay. Human intestinal epithelial cell lines were induced to NEC by lipopolysaccharides (LPSs). LPS-induced cells were transfected with small interfering RNA-HMGB1 and NLRP3 plasmid vector. The mRNA and protein expression of HMGB1, NLRP3, TLR4, NF-κB, caspase 1, IL-1β, and TNF-α were determined by real-time PCR and western blot analysis, respectively. RESULTS The mRNA and protein expression of HMGB1 and NLRP3 in the NEC group was significantly higher than the control group. Inhibition of HMGB1 expression improved intestinal inflammation in newborn NEC rats. The expression of HMGB1, NLRP3, TLR4, NF-κB, and caspase 1 was upregulated in NEC and was weakened after treating with GL. LPS induction to intestinal epithelial cells markedly increased the expression of HMGB1, NLRP3, TLR4, NF-κB, caspase 1, IL-1β, and TNF-α. The knockdown of HMGB1 abolished the increase of expression, whereas further transfection with NLRP3 plasmid vector recovered the increase. CONCLUSION HMGB1 and NLRP3 were all upregulated in the development of NEC. Inhibition on HMGB1 could improve the intestinal inflammation in NEC by inhibiting NLRP3 via TLR4 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Yaqin Tao
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Ping Li
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Juan Yin
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
12
|
Khasbiullina NR, Shilova NV, Navakouski ME, Nokel AY, Knirel YA, Blixt O, Bovin NV. Repertoire of Abs primed by bacteria in gnotobiotic mice. Innate Immun 2018; 24:180-187. [PMID: 29546786 PMCID: PMC6852387 DOI: 10.1177/1753425918763524] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/27/2018] [Accepted: 02/13/2018] [Indexed: 11/16/2022] Open
Abstract
Innate immunity natural Abs (NAbs) execute a number of functions, including protection and surveillance. Despite active research, the stimuli that induce the formation of NAbs are still described only hypothetically. Here, we compared repertoires of anti-glycan Abs in the peripheral blood of mice that received per os various bacteria. The repertoires of Abs of mice primed in this way were compared using a microarray that included about 350 glycans, as well as 150 bacterial polysaccharides. Sterile mice did not possess anti-glycan Abs. Oral inoculation of a single strain or combination of two to four strains of bacteria, as well as putting the animals on short-term nutrition with non-sterile food, did not contribute significantly to the formation of Abs, whereas a single gavage of digested food of non-sterile mice induced the formation of a repertoire close to the natural ones. Interestingly, the priming with polysaccharide Ags (in a composition of the bacterial cell envelope), that is, dominant Ags of bacteria, led to the induction of Abs against typical glycans of mammalian glycoproteins and glycolipids (e.g. Abs of the ABH blood group system) that do not have a structural similarity to the polysaccharides. The results support the importance of early contact with a naïve immune system with microorganisms of the environment to form a normal NAbs repertoire.
Collapse
Affiliation(s)
- Nailia R Khasbiullina
- Shemyakin-Ovchinnikov Institute of
Bioorganic Chemistry, Russian Academy of Sciences, Russia
- Zelinsky Institute of Organic Chemistry,
Russian Academy of Sciences, Russia
| | - Nadezhda V Shilova
- Shemyakin-Ovchinnikov Institute of
Bioorganic Chemistry, Russian Academy of Sciences, Russia
| | - Maxim E Navakouski
- Shemyakin-Ovchinnikov Institute of
Bioorganic Chemistry, Russian Academy of Sciences, Russia
| | - Alexey Yu Nokel
- Shemyakin-Ovchinnikov Institute of
Bioorganic Chemistry, Russian Academy of Sciences, Russia
| | - Yuri A Knirel
- Zelinsky Institute of Organic Chemistry,
Russian Academy of Sciences, Russia
| | - Ola Blixt
- University of Copenhagen, Department of
Chemistry, Denmark
| | - Nicolai V Bovin
- Shemyakin-Ovchinnikov Institute of
Bioorganic Chemistry, Russian Academy of Sciences, Russia
- Auckland University of Technology, New
Zealand
| |
Collapse
|
13
|
Thomas DM, Bell B, Papillon S, Delaplain P, Lim J, Golden J, Bowling J, Wang J, Wang L, Grishin AV, Ford HR. Colonization with Escherichia coli EC 25 protects neonatal rats from necrotizing enterocolitis. PLoS One 2017; 12:e0188211. [PMID: 29190745 PMCID: PMC5708813 DOI: 10.1371/journal.pone.0188211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 11/02/2017] [Indexed: 12/30/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of morbidity and mortality in premature infants; yet its pathogenesis remains poorly understood. To evaluate the role of intestinal bacteria in protection against NEC, we assessed the ability of naturally occurring intestinal colonizer E. coli EC25 to influence composition of intestinal microbiota and NEC pathology in the neonatal rat model. Experimental NEC was induced in neonatal rats by formula feeding/hypoxia, and graded histologically. Bacterial populations were characterized by plating on blood agar, scoring colony classes, and identifying each class by sequencing 16S rDNA. Binding of bacteria to, and induction of apoptosis in IEC-6 enterocytes were examined by plating on blood agar and fluorescent staining for fragmented DNA. E. coli EC 25, which was originally isolated from healthy rats, efficiently colonized the intestine and protected from NEC following introduction to newborn rats with formula at 106 or 108 cfu. Protection did not depend significantly on EC25 inoculum size or load in the intestine, but positively correlated with the fraction of EC25 in the microbiome. Introduction of EC25 did not prevent colonization with other bacteria and did not significantly alter bacterial diversity. EC25 neither induced cultured enterocyte apoptosis, nor protected from apoptosis induced by an enteropathogenic strain of Cronobacter muytjensii. Our results show that E. coli EC25 is a commensal strain that efficiently colonizes the neonatal intestine and protects from NEC.
Collapse
Affiliation(s)
- Debi M Thomas
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Brandon Bell
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Stephanie Papillon
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Patrick Delaplain
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America.,Department of Surgery, University of Southern California, Los Angeles, California, United States of America
| | - Joanna Lim
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Jamie Golden
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America.,Department of Surgery, University of Southern California, Los Angeles, California, United States of America
| | - Jordan Bowling
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America.,Department of Surgery, University of Southern California, Los Angeles, California, United States of America
| | - Jin Wang
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Larry Wang
- Division of Pathology, Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Anatoly V Grishin
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America.,Department of Surgery, University of Southern California, Los Angeles, California, United States of America
| | - Henri R Ford
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States of America.,Department of Surgery, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
14
|
Xing T, Camacho Salazar R, Chen YH. Animal models for studying epithelial barriers in neonatal necrotizing enterocolitis, inflammatory bowel disease and colorectal cancer. Tissue Barriers 2017; 5:e1356901. [PMID: 28795875 DOI: 10.1080/21688370.2017.1356901] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The intestinal epithelial cells line the luminal surface of the entire gastrointestinal tract which is crucial for the absorption of nutrients and prevention of pathogens entering from the external environment. The epithelial barrier plays an important role in organ development, disease pathogenesis, and aging. The major component of an epithelial barrier is the single columnar epithelium and tight junctions. Tight junctions are located at the most apical region of the junctional complex and contain many integral membrane proteins, such as occludin, the claudin family, and junctional adhesion molecules (JAMs). The disruption of intestinal epithelial barriers may lead to several pathophysiological conditions causing malabsorption of nutrition and chronic inflammation. In this review, we provide an update on the alterations of epithelial barriers associated with gut diseases using experimental animal models; we appraise the role of tight junctions in neonatal necrotizing enterocolitis (NEC), inflammatory bowel disease (IBD), and colorectal cancer; we also compare some common features as well as differences and similarities in the pathophysiology of intestinal inflammation in neonatal (NEC) and adult (IBD) gut.
Collapse
Affiliation(s)
- Tiaosi Xing
- a Department of Anatomy and Cell Biology , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| | - Rolando Camacho Salazar
- b Department of Pediatrics , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| | - Yan-Hua Chen
- a Department of Anatomy and Cell Biology , Brody School of Medicine, East Carolina University , Greenville , NC , USA.,b Department of Pediatrics , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| |
Collapse
|
15
|
|
16
|
Bron PA, Kleerebezem M, Brummer RJ, Cani PD, Mercenier A, MacDonald TT, Garcia-Ródenas CL, Wells JM. Can probiotics modulate human disease by impacting intestinal barrier function? Br J Nutr 2017; 117:93-107. [PMID: 28102115 PMCID: PMC5297585 DOI: 10.1017/s0007114516004037] [Citation(s) in RCA: 304] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/29/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022]
Abstract
Intestinal barrier integrity is a prerequisite for homeostasis of mucosal function, which is balanced to maximise absorptive capacity, while maintaining efficient defensive reactions against chemical and microbial challenges. Evidence is mounting that disruption of epithelial barrier integrity is one of the major aetiological factors associated with several gastrointestinal diseases, including infection by pathogens, obesity and diabetes, necrotising enterocolitis, irritable bowel syndrome and inflammatory bowel disease. The notion that specific probiotic bacterial strains can affect barrier integrity fuelled research in which in vitro cell lines, animal models and clinical trials are used to assess whether probiotics can revert the diseased state back to homeostasis and health. This review catalogues and categorises the lines of evidence available in literature for the role of probiotics in epithelial integrity and, consequently, their beneficial effect for the reduction of gastrointestinal disease symptoms.
Collapse
Affiliation(s)
- Peter A. Bron
- NIZO Food Research and BE-Basic Foundation, Kernhemseweg 2, 6718ZB Ede, The Netherlands
| | - Michiel Kleerebezem
- Wageningen University, Host Microbe Interactomics Group, De Elst 1, 6708WD Wageningen, The Netherlands
| | - Robert-Jan Brummer
- Faculty of Medicine and Health, Örebro University, Fakultetsgatan 1, SE-701 82 Örebro, Sweden
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, WELBIO – Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73 B1.73.11, 1200 Brussels, Belgium
| | - Annick Mercenier
- Nestlé Research Center, Nutrition and Health Research, route du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Thomas T. MacDonald
- Barts and The London school of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Clara L. Garcia-Ródenas
- Nestlé Research Center, Nutrition and Health Research, route du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Jerry M. Wells
- Wageningen University, Host Microbe Interactomics Group, De Elst 1, 6708WD Wageningen, The Netherlands
| |
Collapse
|
17
|
Imperial ICVJ, Ibana JA. Addressing the Antibiotic Resistance Problem with Probiotics: Reducing the Risk of Its Double-Edged Sword Effect. Front Microbiol 2016; 7:1983. [PMID: 28018315 PMCID: PMC5156686 DOI: 10.3389/fmicb.2016.01983] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 11/28/2016] [Indexed: 12/27/2022] Open
Abstract
Antibiotic resistance is a global public health problem that requires our attention. Indiscriminate antibiotic use is a major contributor in the introduction of selective pressures in our natural environments that have significantly contributed in the rapid emergence of antibiotic-resistant microbial strains. The use of probiotics in lieu of antibiotic therapy to address certain health conditions in both animals and humans may alleviate these antibiotic-mediated selective pressures. Probiotic use is defined as the actual application of live beneficial microbes to obtain a desired outcome by preventing diseased state or improving general health. Multiple studies have confirmed the beneficial effects of probiotic use in the health of both livestock and humans. As such, probiotics consumption is gaining popularity worldwide. However, concerns have been raised in the use of some probiotics strains that carry antibiotic resistance genes themselves, as they have the potential to pass the antibiotic resistance genes to pathogenic bacteria through horizontal gene transfer. Therefore, with the current public health concern on antibiotic resistance globally, in this review, we underscore the need to screen probiotic strains that are used in both livestock and human applications to assure their safety and mitigate their potential in significantly contributing to the spread of antibiotic resistance genes in our natural environments.
Collapse
Affiliation(s)
- Ivan C V J Imperial
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman Quezon City, Philippines
| | - Joyce A Ibana
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman Quezon City, Philippines
| |
Collapse
|
18
|
Itani T, Ayoub Moubareck C, Melki I, Rousseau C, Mangin I, Butel MJ, Karam Sarkis D. Establishment and development of the intestinal microbiota of preterm infants in a Lebanese tertiary hospital. Anaerobe 2016; 43:4-14. [PMID: 27833033 DOI: 10.1016/j.anaerobe.2016.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/02/2016] [Accepted: 11/06/2016] [Indexed: 12/12/2022]
Abstract
The establishment and development of the intestinal microbiota is known to be associated with profound short- and long-term effects on the health of full-term infants (FTI), but studies are just starting for preterm infants (PTI). The data also mostly come from western countries and little information is available for the Middle East. Here, we determined the composition and dynamics of the intestinal microbiota during the first month of life for PTI (n = 66) and FTI (n = 17) in Lebanon. Fecal samples were collected weekly and analyzed by quantitative PCR (q-PCR) and temporal temperature gradient gel electrophoresis (TTGE). We observed differences in the establishment and composition of the intestinal microbiota between the two groups. q-PCR showed that PTI were more highly colonized by Staphylococcus than FTI in the first three weeks of life; whereas FTI were more highly colonized by Clostridium clusters I and XI. At one month of life, PTI were mainly colonized by facultative anaerobes and a few strict anaerobes, such as Clostridium cluster I and Bifidobacterium. The type of feeding and antibiotic treatments significantly affected intestinal colonization. TTGE revealed low species diversity in both groups and high inter-individual variability in PTI. Our findings show that PTI had altered intestinal colonization with a higher occurrence of potential pathogens (Enterobacter, Clostridium sp) than FTI. This suggests the need for intervention strategies for PTI to modulate their intestinal microbiota and promote their health.
Collapse
Affiliation(s)
- Tarek Itani
- Microbiology Laboratory, School of Pharmacy, Saint-Joseph University, Beirut, Lebanon; Rodolphe Mérieux Laboratory, Beirut, Lebanon.
| | - Carole Ayoub Moubareck
- Microbiology Laboratory, School of Pharmacy, Saint-Joseph University, Beirut, Lebanon; Rodolphe Mérieux Laboratory, Beirut, Lebanon; College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Imad Melki
- Hôtel Dieu de France Hospital, Beirut, Lebanon
| | - Clotilde Rousseau
- EA4065, Hospital University Department, Paris Descartes University, Sorbonne Paris Cité, Paris, France; Microbiology, Saint-Louis Hospital, APHP, Paris, France
| | - Irène Mangin
- EA4065, Hospital University Department, Paris Descartes University, Sorbonne Paris Cité, Paris, France; Laboratoire MIEL, Cnam, Paris, France
| | - Marie-José Butel
- EA4065, Hospital University Department, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Dolla Karam Sarkis
- Microbiology Laboratory, School of Pharmacy, Saint-Joseph University, Beirut, Lebanon; Rodolphe Mérieux Laboratory, Beirut, Lebanon
| |
Collapse
|
19
|
Sellmann C, Jin CJ, Engstler AJ, De Bandt JP, Bergheim I. Oral citrulline supplementation protects female mice from the development of non-alcoholic fatty liver disease (NAFLD). Eur J Nutr 2016; 56:2519-2527. [PMID: 27496089 DOI: 10.1007/s00394-016-1287-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/29/2016] [Indexed: 01/08/2023]
Abstract
PURPOSE Impairments of intestinal barrier function are discussed as risk factors for the development and progression of non-alcoholic fatty liver disease (NAFLD). Studies suggest an association between arginine/citrulline homeostasis and the development of liver damages. Here, the effect of an oral L-citrulline (Cit) supplement on the development of a Western-style diet (WSD)-induced NAFLD was determined in mice. METHODS Female 6- to 8-week-old C57BL/6J mice were either pair-fed a liquid Western-style or control diet (C) ± 2.5 g/kg bodyweight Cit for 6 weeks (C + Cit or WSD + Cit). Indices of liver damage, glucose metabolism, intestinal barrier function and NO synthesis were measured. RESULTS While bodyweight gain was similar between groups, markers of glucose metabolism like fasting blood glucose and HOMA index and markers of liver damage like hepatic triglyceride levels, number of neutrophils and plasminogen activator inhibitor-1 protein levels were significantly lower in WSD + Cit-fed mice when compared to WSD-fed mice only. Protein levels of the tight junction proteins occludin and zonula occludens-1 in duodenum were significantly lower in mice fed a WSD when compared to those fed a WSD + Cit (-~70 and -~60 %, respectively, P < 0.05), whereas portal endotoxin levels, concentration of 3-nitrotyrosine protein adducts in duodenum and toll-like receptor-4 mRNA expression in livers of WSD + Cit-fed mice were markedly lower than in WSD-fed mice (-~43 %, P = 0.056; -~80 and -~48 %, respectively, P < 0.05). CONCLUSION Our data suggest that the protective effects of supplementing Cit on the development of NAFLD in mice are associated with a decreased translocation of endotoxin into the portal vein.
Collapse
Affiliation(s)
- Cathrin Sellmann
- Institute of Nutritional Sciences, SD Model Systems of Molecular Nutrition, Friedrich-Schiller University Jena, Dornburger Str. 29, 07743, Jena, Germany
| | - Cheng Jun Jin
- Institute of Nutritional Sciences, SD Model Systems of Molecular Nutrition, Friedrich-Schiller University Jena, Dornburger Str. 29, 07743, Jena, Germany
| | - Anna Janina Engstler
- Institute of Nutritional Sciences, SD Model Systems of Molecular Nutrition, Friedrich-Schiller University Jena, Dornburger Str. 29, 07743, Jena, Germany
| | - Jean-Pascal De Bandt
- Nutrition Biology Laboratory EA4466, Faculty of Pharmacy, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Clinical Chemistry Department, Paris Centre University Hospitals, APHP, Paris, France
| | - Ina Bergheim
- Institute of Nutritional Sciences, SD Model Systems of Molecular Nutrition, Friedrich-Schiller University Jena, Dornburger Str. 29, 07743, Jena, Germany.
| |
Collapse
|
20
|
Stirling KM, Hussain N, Sanders MM, Campbell W. Association between maternal genital mycoplasma colonization and histologic chorioamnionitis in preterm births. J Neonatal Perinatal Med 2016; 9:201-209. [PMID: 27197925 DOI: 10.3233/npm-16915059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
OBJECTIVE Genital mycoplasmas (GMs) can be associated with chorioamnionitis and preterm birth, but are viewed as commensal organisms with low virulence. We sought to determine if cervical GM colonization is associated with histologic chorioamnionitis independent of infection with other bacteria. STUDY DESIGN Retrospective study of patients who delivered preterm, had cervical cultures for GMs, and placental cultures for bacteria other than GM. GM positive patients were compared to GM negative patients. Histologic grading of placentas was compared between GM negative patients with negative placental cultures (Group 1), GM positive patients with negative placental cultures (Group 2), GM negative patients with positive placental cultures (Group 3) and GM positive patients with positive placental cultures (Group 4). RESULTS GM positive patients were less likely than GM negative patients to have placental cultures positive for other bacteria (39% versus 47%, P = 0.0071). Group 2 had higher rates of membrane inflammation compared to Group 1 (p = 0.0079), and no significant difference in rates of membrane inflammation compared to Groups 3 or 4 (p = 0.36, p = 0.18). GM positivity was independently associated with increased membrane inflammation and decreased inflammation in the chorionic plate. CONCLUSIONS GM colonization is associated decreased inflammation of the chorionic plate, and increased inflammation of the membranes.
Collapse
Affiliation(s)
- K M Stirling
- Department of Maternal-Fetal Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - N Hussain
- Department of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA
| | - M M Sanders
- Department of Pathology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - W Campbell
- Department of Maternal-Fetal Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
21
|
Abstract
The Cronobacter group of pathogens, associated with severe and potentially life-threatening diseases, until recently were classified as a single species, Enterobacter sakazakii. The group was reclassified in 2007 into the genus Cronobacter as a member of the Enterobacteriaceae. This chapter outlines the history behind the epidemiology, analyzes how our understanding of these bacteria has evolved, and highlights the clinical significance the Cronobacter spp. have for neonatal and elderly patient populations and treatment of the associated infections.
Collapse
|
22
|
Cassir N, Simeoni U, La Scola B. Gut microbiota and the pathogenesis of necrotizing enterocolitis in preterm neonates. Future Microbiol 2016; 11:273-92. [PMID: 26855351 DOI: 10.2217/fmb.15.136] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Necrotizing enterocolitis (NEC) remains a devastating intestinal disease in preterm neonates. In this population, disruption of the gut microbiota development, mainly due to organ immaturity, antibiotic use and hospital microbial environment, plays a key role in the pathogenesis of NEC. This gut dysbiosis has been associated with opportunistic pathogens overgrowth, expression of virulence factors, altered metabolic functions and inflammatory dysregulated responses. In this review, we provide an updated summary of the host and gut microbiota interactions during the formative early life. We also explore the key determinants of gut dysbiosis in preterm neonates with NEC. Finally, we discuss the promising role of bacteriotherapy in the management of NEC, the aim being to shape or restore the beneficial gut bacterial communities.
Collapse
Affiliation(s)
- Nadim Cassir
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE), UM63, CNRS 7278, IRD 198, INSERM U1095, Facultés de Médecine et de Pharmacie, Aix-Marseille Université, Marseille, France
| | - Umberto Simeoni
- Service de Pédiatrie, Centre Hospitalier Universitaire Vaudois, Lausanne, Suisse, Switzerland
| | - Bernard La Scola
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE), UM63, CNRS 7278, IRD 198, INSERM U1095, Facultés de Médecine et de Pharmacie, Aix-Marseille Université, Marseille, France
| |
Collapse
|
23
|
Dollings MC, Brown L. An Integrated Review of Intestinal Microbiota in the Very Premature Infant. Neonatal Netw 2016; 35:204-216. [PMID: 27461199 DOI: 10.1891/0730-0832.35.4.204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
BACKGROUND The intestinal microbiota is important for optimal intestinal function and protection against infection. Disruption of the intestinal microbiota has been linked to necrotizing enterocolitis (NEC) and late-onset sepsis (LOS). OBJECTIVES To determine what is known about the intestinal microbiota of very premature infants. We describe the relationship of the intestinal microbiota to NEC and LOS and provide an overview of the effect of environmental factors. METHODS A review was conducted using PubMed/MEDLINE from 2008 to 2015 to examine what is currently known about the intestinal microbiota of very premature infants. DISCUSSION The intestinal bacteria are low in diversity and generally contain a predominance of potentially pathogenic bacteria. Evidence suggests that these bacteria play a role in NEC as well as LOS through translocation. Environmental factors such as mode of delivery, use of antibiotics, and type of feeding may also contribute to the development of the intestinal microbiota.
Collapse
Affiliation(s)
- Melissa C Dollings
- Virginia Commonwealth University, 9631 Raven Wing Drive, Chesterfield, VA 23832, USA
| | | |
Collapse
|
24
|
Franklin AL, Said M, Cappiello CD, Gordish-Dressman H, Tatari-Calderone Z, Vukmanovic S, Rais-Bahrami K, Luban NLC, Devaney JM, Sandler AD. Are Immune Modulating Single Nucleotide Polymorphisms Associated with Necrotizing Enterocolitis? Sci Rep 2015; 5:18369. [PMID: 26670709 PMCID: PMC4680983 DOI: 10.1038/srep18369] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency. The purpose of this study is to determine if functional single nucleotide polymorphisms (SNPs) in immune-modulating genes pre-dispose infants to NEC. After Institutional Review Board approval and parental consent, buccal swabs were collected for DNA extraction. TaqMan allelic discrimination assays and BglII endonuclease digestion were used to genotype specific inflammatory cytokines and TRIM21. Statistical analysis was completed using logistic regression. 184 neonates were analyzed in the study. Caucasian neonates with IL-6 (rs1800795) were over 6 times more likely to have NEC (p = 0.013; OR = 6.61, 95% CI 1.48–29.39), and over 7 times more likely to have Stage III disease (p = 0.011; OR = 7.13, (95% CI 1.56–32.52). Neonates with TGFβ-1 (rs2241712) had a decreased incidence of NEC-related perforation (p = 0.044; OR = 0.28, 95% CI: 0.08–0.97) and an increased incidence of mortality (p = 0.049; OR = 2.99, 95% CI: 1.01 – 8.86). TRIM21 (rs660) was associated with NEC-related intestinal perforation (p = 0.038; OR = 4.65, 95% CI 1.09–19.78). In premature Caucasian neonates, the functional SNP IL-6 (rs1800795) is associated with both the development and increased severity of NEC. TRIM21 (rs660) and TGFβ-1 (rs2241712) were associated with NEC- related perforation in all neonates in the cohort. These findings suggest a possible genetic role in the development of NEC.
Collapse
Affiliation(s)
- Ashanti L Franklin
- Division of General and Thoracic Surgery, Children's National Health System, 111 Michigan Ave NW, Washington, DC 20010
| | - Mariam Said
- Division of Neonatology, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Clint D Cappiello
- Division of General and Thoracic Surgery, Children's National Health System, 111 Michigan Ave NW, Washington, DC 20010
| | - Heather Gordish-Dressman
- Children's Research Institute, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Zohreh Tatari-Calderone
- Sheikh Zayed Institute, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Stanislav Vukmanovic
- Sheikh Zayed Institute, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Khodayar Rais-Bahrami
- Division of Neonatology, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Naomi L C Luban
- Department Laboratory Medicine, Children's National Health System Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Joseph M Devaney
- Department of Genetic Medicine, Children's National Health System, 111 Michigan Ave NW, Washington, DC 20010
| | - Anthony D Sandler
- Division of General and Thoracic Surgery, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| |
Collapse
|
25
|
Ditzenberger G. Nutritional support for premature infants in the neonatal intensive care unit. Crit Care Nurs Clin North Am 2015; 26:181-98. [PMID: 24878205 DOI: 10.1016/j.ccell.2014.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nutritional support for premature infants in the neonatal intensive care unit setting is complex. Such infants have conditions unique to this period of the lifespan requiring specialized care management, both of which may impede the provision of adequate nutrition to support basal metabolic needs. Premature infants require optimum nutritional intake to support rapid growth during a time when they are not fully capable of tolerating it. This article reviews developmental anatomy, physiology, and the effect of premature delivery by systems; the challenges of providing adequate nutrition; and current evidence-based strategies to provide nutrition for premature infants during hospitalization.
Collapse
|
26
|
Dysbiosis of Intestinal Microbiota Associated With Inflammation Involved in the Progression of Acute Pancreatitis. Pancreas 2015; 44:868-75. [PMID: 25931253 DOI: 10.1097/mpa.0000000000000355] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To evaluate alterations of the intestinal bacteria and its associations with the inflammation in acute pancreatitis (AP). METHODS A multihospital prospective clinical study was conducted, and a total of 108 participants were enrolled in our study, including 44 with severe AP (SAP), 32 with mild AP (MAP), and 32 healthy volunteers. The structure of intestinal microbiota, 10 predominant bacteria, plasma endotoxin, and serum cytokines were investigated by polymerase chain reaction-denaturing gradient gel electrophoresis, real-time quantitative polymerase chain reaction, Limulus amebocyte lysate tests, and enzyme-linked immunosorbent assays, respectively. RESULTS Dramatic alterations in the predominant fecal microbiota were observed in most of both MAP and SAP patients. In addition, the rates of the multiorgan failures and infectious complications in the patients with SAP with altered intestinal microbiota were significantly higher than in those whose intestinal microbiota remained unaltered. Enterococcus increased and Bifidobacterium decreased in the patients with SAP compared to the patients with MAP. Serum IL-6 were positively correlated with Enterobacteriaceae and Enterococcus and negatively correlated with Bifidobacterium, whereas plasma endotoxin positively correlated with Enterococcus (P < 0.05). CONCLUSIONS The intestinal bacteria most frequently altered in both the patients with MAP and those with SAP significantly correlated with inflammation, which indicated that the intestinal microbiota may be involved in the progression of AP.
Collapse
|
27
|
McElroy SJ, Castle SL, Bernard JK, Almohazey D, Hunter CJ, Bell BA, Al Alam D, Wang L, Ford HR, Frey MR. The ErbB4 ligand neuregulin-4 protects against experimental necrotizing enterocolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2768-78. [PMID: 25216938 DOI: 10.1016/j.ajpath.2014.06.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 06/05/2014] [Accepted: 06/13/2014] [Indexed: 12/30/2022]
Abstract
Necrotizing enterocolitis (NEC) affects up to 10% of premature infants, has a mortality of 30%, and can leave surviving patients with significant morbidity. Neuregulin-4 (NRG4) is an ErbB4-specific ligand that promotes epithelial cell survival. Thus, this pathway could be protective in diseases such as NEC, in which epithelial cell death is a major pathologic feature. We sought to determine whether NRG4-ErbB4 signaling is protective in experimental NEC. NRG4 was used i) in the newborn rat formula feeding/hypoxia model; ii) in a recently developed model in which 14- to 16-day-old mice are injected with dithizone to induce Paneth cell loss, followed by Klebsiella pneumoniae infection to induce intestinal injury; and iii) in bacterially infected IEC-6 cells in vitro. NRG4 reduced NEC incidence and severity in the formula feed/hypoxia rat model. It also reduced Paneth cell ablation-induced NEC and prevented dithizone-induced Paneth cell loss in mice. In vitro, cultured ErbB4(-/-) ileal epithelial enteroids had reduced Paneth cell markers and were highly sensitive to inflammatory cytokines. Furthermore, NRG4 blocked, through a Src-dependent pathway, Cronobacter muytjensii-induced IEC-6 cell apoptosis. The potential clinical relevance of these findings was demonstrated by the observation that NRG4 and its receptor ErbB4 are present in human breast milk and developing human intestine, respectively. Thus, NRG4-ErbB4 signaling may be a novel pathway for therapeutic intervention or prevention in NEC.
Collapse
Affiliation(s)
- Steven J McElroy
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | - Shannon L Castle
- Division of Pediatric Surgery, Department of Surgery, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Jessica K Bernard
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California
| | - Dana Almohazey
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California
| | - Catherine J Hunter
- Departments of Surgery and Pediatrics, Ann and Robert H. Lurie Children's Hospital, Northwestern University, Chicago, Illinois
| | - Brandon A Bell
- Division of Pediatric Surgery, Department of Surgery, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Denise Al Alam
- Division of Pediatric Surgery, Department of Surgery, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Larry Wang
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Henri R Ford
- Division of Pediatric Surgery, Department of Surgery, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Mark R Frey
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California; Department of Biochemistry and Molecular Biology, University of Southern California Keck School of Medicine, Los Angeles, California.
| |
Collapse
|
28
|
Short SS, Papillon S, Berel D, Ford HR, Frykman PK, Kawaguchi A. Late onset of necrotizing enterocolitis in the full-term infant is associated with increased mortality: results from a two-center analysis. J Pediatr Surg 2014; 49:950-3. [PMID: 24888841 PMCID: PMC4370287 DOI: 10.1016/j.jpedsurg.2014.01.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 01/27/2014] [Indexed: 12/30/2022]
Abstract
PURPOSE The effect of timing of onset of necrotizing enterocolitis (NEC) on outcomes has not been determined for the full-term infant. In this study we aimed to characterize the full-term NEC population and to evaluate onset of NEC. METHODS We performed a two-center retrospective review of all full-term infants (≥ 37weeks) with a diagnosis of NEC between 1990 and 2012. Patients were identified by ICD-9 and age. Early onset for NEC was ≤7days and late onset after 7days of life. Demographics, comorbidities, maternal factors, clinical factors, surgical intervention, complications, and mortality were evaluated. Wilcoxon's test was performed on continuous variables and Fisher's exact test on categorical data. A p-value<0.05 was considered significant. Univariate outcomes with a p-value<0.1 were selected for multivariable analysis. RESULTS Thirty-nine patients (24 boys, 15 girls) with median EGA of 39weeks were identified. Overall mortality was 18%. Univariate predictors of mortality included congenital heart disease and placement of an umbilical artery (UA) catheter. Multivariate analysis revealed late onset of NEC to be an independent predictor of mortality (OR 90.8, 95% CI 2.6-3121). CONCLUSION Full-term infants who develop NEC after 7days of life, have congenital heart disease, and/or need UA catheterization have increased mortality.
Collapse
Affiliation(s)
- Scott S. Short
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, and the Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA, Division of Pediatric Surgery, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephanie Papillon
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, and the Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Dror Berel
- Division of Pediatric Surgery, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Henri R. Ford
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, and the Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Philip K. Frykman
- Division of Pediatric Surgery, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Akemi Kawaguchi
- Division of Pediatric Surgery, Children's Hospital Los Angeles, and the Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
29
|
Necrotizing enterocolitis: have we made any progress in reducing the risk? Adv Neonatal Care 2013; 13:317-24; quiz 325-6. [PMID: 24042135 DOI: 10.1097/anc.0b013e31829a872c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Necrotizing enterocolitis (NEC) is a disease affecting premature infants with potentially devastating complications in the neonatal intensive care unit (NICU). Inadequate knowledge regarding the underlying pathophysiology of this disease has contributed to the minimal progress made in decreasing the incidence and severity of NEC. Because of an improved survival rate of the most immature infants, the number of diagnosed cases of NEC is anticipated to increase. Unfortunately, decades of research have failed to reduce the risk or improve the prognosis of NEC, magnifying the need for risk-reduction strategies for decreasing significant NEC-related morbidity and mortality. Advancements in our understanding of this disease process have facilitated the development of several strategies that have been investigated in the risk reduction of NEC. This article discusses the pathophysiology and causes of NEC and presents strategies investigated regarding risk reduction of this devastating disease.
Collapse
|
30
|
Host defense against common early life-threatening infections. Clin Dev Immunol 2013; 2013:350808. [PMID: 24093045 PMCID: PMC3777204 DOI: 10.1155/2013/350808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 06/23/2013] [Indexed: 11/21/2022]
|
31
|
Cronobacter: an emerging opportunistic pathogen associated with neonatal meningitis, sepsis and necrotizing enterocolitis. J Perinatol 2013; 33:581-5. [PMID: 23538645 DOI: 10.1038/jp.2013.26] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 01/24/2013] [Accepted: 02/11/2013] [Indexed: 11/08/2022]
Abstract
Members of the genus Cronobacter are an emerging group of opportunist Gram-negative pathogens. This genus was previously thought to be a single species, called Enterobacter sakazakii. Cronobacter spp. typically affect low-birth-weight neonates, causing life-threatening meningitis, sepsis and necrotizing enterocolitis. Outbreaks of disease have been associated with contaminated infant formula, although the primary environmental source remains elusive. Advanced understanding of these bacteria and better classification has been obtained by improved detection techniques and genomic analysis. Research has begun to characterize the virulence factors and pathogenic potential of Cronobacter. Investigations into sterilization techniques and protocols for minimizing the risk of contamination have been reviewed at national and international forums. In this review, we explore the clinical impact of Cronobacter neonatal and pediatric infections, discuss virulence and pathogenesis, and review prevention and treatment strategies.
Collapse
|
32
|
Martin CR, Bellomy M, Allred EN, Fichorova RN, Leviton A. Systemic inflammation associated with severe intestinal injury in extremely low gestational age newborns. Fetal Pediatr Pathol 2013; 32:222-34. [PMID: 23002960 PMCID: PMC4201234 DOI: 10.3109/15513815.2012.721477] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
To define the role of systemic inflammation in infants with intestinal perforation (IP) and necrotizing enterocolitis (NEC), we measured 25 blood protein concentrations on days 1, 7 and 14 in 939 infants born before 28 weeks' gestation. On days 7 and 14, infants with NEC had elevated levels of C-reactive protein (CRP), serum amyloid A (SAA), IL-6 and IL-8. Infants with IP had elevated levels of CRP and insulin growth factor binding protein-1 on day 7 and elevated CRP, SAA, TNF-receptor-2 and matrix metalloproteinase-9 levels on day 14. A better understanding of systemic inflammation might help prevent and treat these disorders.
Collapse
Affiliation(s)
- Camilia R Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | | | | | | | | |
Collapse
|
33
|
Neonatal sepsis due to coagulase-negative staphylococci. Clin Dev Immunol 2013; 2013:586076. [PMID: 23762094 PMCID: PMC3674645 DOI: 10.1155/2013/586076] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/27/2013] [Accepted: 04/27/2013] [Indexed: 02/06/2023]
Abstract
Neonates, especially those born prematurely, are at high risk of morbidity and mortality from sepsis. Multiple factors, including prematurity, invasive life-saving medical interventions, and immaturity of the innate immune system, put these infants at greater risk of developing infection. Although advanced neonatal care enables us to save even the most preterm neonates, the very interventions sustaining those who are hospitalized concurrently expose them to serious infections due to common nosocomial pathogens, particularly coagulase-negative staphylococci bacteria (CoNS). Moreover, the health burden from infection in these infants remains unacceptably high despite continuing efforts. In this paper, we review the epidemiology, immunological risk factors, diagnosis, prevention, treatment, and outcomes of neonatal infection due to the predominant neonatal pathogen CoNS.
Collapse
|
34
|
Antenatal ureaplasma infection impairs development of the fetal ovine gut in an IL-1-dependent manner. Mucosal Immunol 2013; 6:547-56. [PMID: 23149664 DOI: 10.1038/mi.2012.97] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Ureaplasma infection of the amniotic cavity is associated with adverse postnatal intestinal outcomes. We tested whether interleukin-1 (IL-1) signaling underlies intestinal pathology following ureaplasma exposure in fetal sheep. Pregnant ewes received intra-amniotic injections of ureaplasma or culture media for controls at 3, 7, and 14 d before preterm delivery at 124 d gestation (term 150 d). Intra-amniotic injections of recombinant human interleukin IL-1 receptor antagonist (rhIL-1ra) or saline for controls were given 3 h before and every 2 d after Ureaplasma injection. Ureaplasma exposure caused fetal gut inflammation within 7 d with damaged villus epithelium and gut barrier loss. Proliferation, differentiation, and maturation of enterocytes were significantly reduced after 7 d of ureaplasma exposure, leading to severe villus atrophy at 14 d. Inflammation, impaired development and villus atrophy of the fetal gut was largely prevented by intra-uterine rhIL-1ra treatment. These data form the basis for a clinical understanding of the role of ureaplasma in postnatal intestinal pathologies.
Collapse
|
35
|
Grishin A, Papillon S, Bell B, Wang J, Ford HR. The role of the intestinal microbiota in the pathogenesis of necrotizing enterocolitis. Semin Pediatr Surg 2013; 22:69-75. [PMID: 23611609 PMCID: PMC3647029 DOI: 10.1053/j.sempedsurg.2013.01.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Development of necrotizing enterocolitis (NEC) requires a susceptible host, typically a premature infant or an infant with congenital heart disease, enteral feedings and bacterial colonization. Although there is little doubt that microbes are critically involved in the pathogenesis of NEC, the identity of specific causative pathogens remains elusive. Unlike established normal adult gut microbiota, which is quite complex, uniform, and stable, early postnatal bacterial populations are simple, diverse, and fluid. These properties complicate studies aimed at elucidating characteristics of the gut microbiome that may play a role in the pathogenesis of NEC. A broad variety of bacterial, viral, and fungal species have been implicated in both clinical and experimental NEC. Frequently, however, the same species have also been found in physiologically matched healthy individuals. Clustered outbreaks of NEC, in which the same strain of a suspected pathogen is detected in several patients suggest, but do not prove, a causative relationship between the specific pathogen and the disease. Studies in Cronobacter sakazakii, the best characterized NEC pathogen, have demonstrated that virulence is not a property of a bacterial species as a whole, but rather a characteristic of certain strains, which may explain why the same species can be pathogenic or non-pathogenic. The fact that a given microbe may be innocuous in a full-term, yet pathogenic in a pre-term infant has led to the idea of opportunistic pathogens in NEC. Progress in understanding the infectious nature of NEC may require identifying specific pathogenic strains and unambiguously establishing their virulence in animal models.
Collapse
|
36
|
Däbritz J, Jenke A, Wirth S, Foell D. Fecal phagocyte-specific S100A12 for diagnosing necrotizing enterocolitis. J Pediatr 2012; 161:1059-64. [PMID: 22796048 DOI: 10.1016/j.jpeds.2012.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/14/2012] [Accepted: 06/01/2012] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To determine whether longitudinal measurements of fecal S100A12, a fecal marker of intestinal inflammation, can identify very low birth weight infants at risk for necrotizing enterocolitis (NEC). STUDY DESIGN This prospective study included 145 preterm infants with birth weight <1500 g. Meconium and stool samples (n = 843) were collected prospectively on alternate days for 4 weeks, and fecal S100A12 and calprotectin were measured by enzyme-linked immunosorbent assay. RESULTS Eighteen patients (12.4%) developed NEC. Gestational age and birth weight were significantly lower in the patients with NEC compared with unaffected reference infants. Fecal S100A12 levels were significantly higher in patients with severe NEC at onset of disease and also, in contrast to fecal calprotectin, at 4-10 days before onset of NEC compared with unaffected reference infants (ideal cutoff value, 65 μg/kg; sensitivity, 0.76; specificity, 0.56). CONCLUSIONS Fecal S100A12 level may be a helpful marker for predicting disease severity and early risk assessment for subsequent development of NEC. However, the use of fecal S100A12 as a predictive biomarker for NEC in very low birth weight infants may be limited due to a high interindividual and intraindividual variability in S100A12 fecal excretion.
Collapse
Affiliation(s)
- Jan Däbritz
- Institute of Immunology, University Hospital Münster, Münster, Germany.
| | | | | | | |
Collapse
|
37
|
Arboleya S, Solís G, Fernández N, de los Reyes-Gavilán CG, Gueimonde M. Facultative to strict anaerobes ratio in the preterm infant microbiota: a target for intervention? Gut Microbes 2012; 3:583-8. [PMID: 22922559 PMCID: PMC3495798 DOI: 10.4161/gmic.21942] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
During recent years there has been an increasing interest on the development of strategies for modulating the process of microbiota establishment in preterm infants. For successfully developing of such strategies, a detailed knowledge of the microbiota establishment process in these infants is needed. In a previous study we evidenced clear alterations in the process of microbiota establishment in preterm newborns when compared with a control group of full-term breast-fed infants. Here we have analyzed these data more in depth, corroborating a reduced proportion of strict anaerobes with respect to facultatives in the fecal microbiota of preterm infants. The potential benefits, as well as the side effects, of strategies aimed at counterbalancing this alteration in the facultative to strict anaerobes ratio are discussed in this addendum.
Collapse
Affiliation(s)
- Silvia Arboleya
- Department of Microbiology and Biochemistry; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa, Asturias, Spain
| | - Gonzalo Solís
- Paediatrics Service; Hospital Universitario Central de Asturias; Servicio de Salud del Principado de Asturias; Oviedo, Asturias, Spain
| | - Nuria Fernández
- Paediatrics Service; Hospital Cabueñes; Servicio de Salud del Principado de Asturias; Gijón, Asturias, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa, Asturias, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa, Asturias, Spain,Correspondence to: Miguel Gueimonde,
| |
Collapse
|
38
|
McElroy SJ, Underwood MA, Sherman MP. Paneth cells and necrotizing enterocolitis: a novel hypothesis for disease pathogenesis. Neonatology 2012; 103:10-20. [PMID: 23006982 PMCID: PMC3609425 DOI: 10.1159/000342340] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/02/2012] [Indexed: 12/18/2022]
Abstract
Current models of necrotizing enterocolitis (NEC) propose that intraluminal microbes destroy intestinal mucosa and activate an inflammatory cascade that ends in necrosis. We suggest an alternate hypothesis wherein NEC is caused by injury to Paneth cells (PCs) in the intestinal crypts. PCs are specialized epithelia that protect intestinal stem cells from pathogens, stimulate stem cell differentiation, shape the intestinal microbiota, and assist in repairing the gut. Our novel model of NEC uses neonatal mice and ablates PCs followed by enteral infection. We contrast this model with other animal examples of NEC and the clinical disease. Selective destruction of PCs using dithizone likely releases tumor necrosis factor-α and other inflammatory mediators. We propose that this event produces inflammation in the submucosa, generates platelet-activating factor, and induces a coagulopathy. The role of PCs in NEC is consistent with the onset of disease in preterm infants after a period of PC-related maturation, the central role of PCs in crypt-related homeostasis, the anatomic location of pneumatosis intestinalis close to the crypts, and the proximity of PCs to occluded blood vessels that cause coagulation necrosis of the intestinal villi. We offer this hypothesis to promote new thoughts about how NEC occurs and its potential prevention.
Collapse
Affiliation(s)
- Steven J McElroy
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | | | | |
Collapse
|
39
|
Abstract
OBJECTIVE To determine whether an association exists between antenatal antibiotic exposure and incidence of necrotizing enterocolitis (NEC) in low birth weight infants. STUDY DESIGN A retrospective case-control study was performed on all infants with a diagnosis of NEC born at our institition between 1988 and 2006. Medical histories of all infants with a diagnosis of NEC ≥Bell's stage IIA and matched controls without NEC were reviewed. Maternal and neonatal characteristics were compared using the Mantel-Haenszel chi-square procedure, and logistic regression models were constructed to account for confounding. RESULT Clinical data for 97 matched pairs were analyzed. The adjusted odds ratio (OR) for antenatal exposure to ampicillin was significantly greater for infants who developed NEC (OR 2.3, 95% confidence interval 1.1, 4.8, P=0.003) than for control infants. CONCLUSION Infants who developed NEC were more likely to have a history of in utero exposure to ampicillin in the immediate antepartum period than infants who did not develop NEC.
Collapse
|
40
|
Abstract
Necrotizing enterocolitis (NEC) is the most common acquired gastrointestinal disease of premature neonates and is a serious cause of morbidity and mortality. NEC is one of the leading causes of death in neonatal intensive care units. Surgical treatment is necessary in patients whose disease progresses despite medical therapy. Surgical options include peritoneal drainage and laparotomy, with studies showing no difference in outcome related to approach. Survivors, particularly those requiring surgery, face serious sequelae.
Collapse
|
41
|
Arboleya S, González S, Salazar N, Ruas-Madiedo P, de los Reyes-Gavilán CG, Gueimonde M. Development of probiotic products for nutritional requirements of specific human populations. Eng Life Sci 2012. [DOI: 10.1002/elsc.201100129] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Sonia González
- Department of Functional Biology; University of Oviedo; Oviedo; Asturias; Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| |
Collapse
|
42
|
Li N, Ma L, Liu X, Shaw L, Calzi SL, Grant MB, Neu J. Arginyl-glutamine dipeptide or docosahexaenoic acid attenuates hyperoxia-induced small intestinal injury in neonatal mice. J Pediatr Gastroenterol Nutr 2012; 54:499-504. [PMID: 22020559 PMCID: PMC3749514 DOI: 10.1097/mpg.0b013e3182330867] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND OBJECTIVE Supplementation studies of glutamine, arginine, and docosahexaenoic acid (DHA) have established the safety of each of these nutrients in neonates; however, the potential for a more stable and soluble dipeptide, arginyl-glutamine (Arg-Gln) or DHA with anti-inflammatory properties, to exert benefits on hyperoxia-induced intestinal injury has not been investigated. Arg-Gln dipeptide has been shown to prevent retinal damage in a rodent model of oxygen-induced injury. The objective of the present study was to investigate whether Arg-Gln dipeptide or DHA could also attenuate markers of injury and inflammation to the small intestine in this same model. METHODS Seven-day-old mouse pups were placed with their dams in 75% oxygen for 5 days. After 5 days of hyperoxic exposure (P7-P12), pups were removed from hyperoxia and allowed to recover in atmospheric conditions for 5 days (P12-P17). Mouse pups received Arg-Gln (5g·kg·day) or DHA (5g·kg·day) or vehicle orally started on P12 through P17. Distal small intestine (DSI) histologic changes, myeloperoxidase (MPO), lactate dehydrogenase (LDH), inflammatory cytokines, and tissue apoptosis were evaluated. RESULTS Hyperoxic mice showed a greater distortion of overall villus structure and with higher injury score (P<0.05). Arg-Gln dipeptide and DHA supplementation groups were more similar to the room air control group. Supplementation of Arg-Gln or DHA reduced hyperoxia-induced MPO activity (P<0.05). Supplementation of Arg-Gln or DHA returned LDH activity to the levels of control. Hyperoxia induced apoptotic cell death in DSIs, and both Arg-Gln and DHA reversed this effect (P<0.05). CONCLUSIONS Supplementation with either Arg-Gln or DHA may limit some inflammatory and apoptotic processes involved in hyperoxic-induced intestinal injury in neonatal mice.
Collapse
Affiliation(s)
- Nan Li
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Liya Ma
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Xueyan Liu
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Lynn Shaw
- Department of Pharmacology, University of Florida, Gainesville, FL
| | - Sergio Li Calzi
- Department of Pharmacology, University of Florida, Gainesville, FL
| | - Maria B. Grant
- Department of Pharmacology, University of Florida, Gainesville, FL
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL
| |
Collapse
|
43
|
Neonatal T-cell maturation and homing receptor responses to Toll-like receptor ligands differ from those of adult naive T cells: relationship to prematurity. Pediatr Res 2012; 71:136-43. [PMID: 22258123 PMCID: PMC3394681 DOI: 10.1038/pr.2011.26] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Inflammation and infection are associated with premature birth and with activation of the fetal immune system. We hypothesized that exposure to microbial Toll-like receptor (TLR) ligands plays an important role in neonatal T-cell maturation and that early exposure to microbial products may result in early T-cell maturation and a tendency for these matured effector cells to change their homing receptor patterns. RESULTS Expression of the CD45RO marker was induced in term neonatal T cells after in vitro exposure to TLR ligands for 7 days. Interestingly, naive T cells from adult blood were unaffected by TLR ligand exposure. In addition, neonatal T cells had more cells with decreased expression of the α4β7 integrins and increased expression of CCR4 after in vitro exposure of TLR ligands-similar to the expression of these molecules in adult naive T cells. DISCUSSION These findings are relevant for the understanding of neonatal T-cell maturation and may contribute to our understanding of multiorgan inflammatory complications of prematurity. METHODS Cord blood was obtained from term and preterm infants. Using flow cytometry, we identified a mature (CD45RO(+)) phenotype in preterm infant cord blood (CB) T cells that had decreased expression of the α4β7 integrins and increased expression of the C-C chemokine receptor 4 (CCR4) as compared with term infant CB.
Collapse
|
44
|
Prematurity does not markedly affect intestinal sensitivity to endotoxins and feeding in pigs. Br J Nutr 2011; 108:672-81. [DOI: 10.1017/s0007114511006404] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Preterm neonates show enhanced sensitivity to nutrient maldigestion and bacteria-mediated gut inflammatory disorders, such as necrotising enterocolitis (NEC). We hypothesised that preterm birth increases the sensitivity of intestinal nutrient absorption to endotoxins and that feeding after birth reduces this response. Hence, we investigated the postnatal development of nutrient digestive and absorptive capacity in the preterm and term pig intestine, and its responsiveness to endotoxins. Pigs were delivered by caesarean section at preterm (n 20) or term (n 17) gestation, and the small intestine was collected at birth or after 2 d of colostrum feeding, followed by ex vivo stimulation with lipopolysaccharide endotoxins and mixed gut contents collected from pigs with NEC. Brush border enzyme activities were reduced in newborn preterm v. term pigs (39–45 % reduction, P < 0·05), but normalised after 2 d of feeding. Ex vivo leucine and glucose uptake increased with prenatal age. Bacterial stimulation reduced the nutrient uptake similarly at birth and after 2 d in preterm and term pigs (23–41 % reduction, P < 0·05), whereas IL-6 and TNF-α expression was stimulated only at birth. Toll-like receptor-4 expression increased markedly at day 2 for preterm and term pigs (22–33-fold, P < 0·05) but with much lower expression levels in newborn preterm pigs (approximately 95 %, P < 0·01). In conclusion, digestive and absorptive functions mature in the prenatal period, but are similarly affected by postnatal feeding and bacterial exposure in both preterm and term pigs. Nutrient maldigestion may contribute to NEC development, while a prematurity-related hyper-responsiveness to endotoxins could be less important, at least in pigs.
Collapse
|
45
|
Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV. Role of neutrophils and macrophages in the pathogenesis of necrotizing enterocolitis caused by Cronobacter sakazakii. J Surg Res 2011; 172:18-28. [PMID: 21601887 DOI: 10.1016/j.jss.2011.04.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/17/2011] [Accepted: 04/07/2011] [Indexed: 01/25/2023]
Abstract
BACKGROUND Cronobacter sakazakii (CS) is a highly virulent gram-negative opportunistic pathogen that has been implicated in clinical outbreaks of necrotizing enterocolitis (NEC). The role of mucosal immune cells in CS infection is not well understood. In this study, we sought to elucidate the role of neutrophils (polymorphonuclear leukocytes; PMNs) and macrophages in the pathogenesis of NEC induced by CS using a novel newborn mouse model. MATERIALS AND METHODS PMNs and macrophages were depleted in newborn mice using Gr-1 antibody and carrageenan, respectively, and then infected with 10(3) CFU of CS. The development of NEC in these mice was assessed by a pathologist based on the morphologic changes in the intestine. Cytokine production was determined in the serum and intestinal homogenates of infected mice by enzyme-linked immunosorbent assay (ELISA). Inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production was determined by flow cytometry and Greiss method, respectively. RESULTS Depletion of PMNs and macrophages in newborn mice led to increased recruitment of dendritic cells (DCs) in the intestine compared with wild-type mice upon infection with CS. PMN- and macrophage-depleted mice showed increased bacterial load, production of pro-inflammatory cytokines, iNOS expression, and NO production in the intestines in comparison to wild-type mice fed with CS. In addition, depletion of PMNs and macrophages prior to infection in mice resulted in severe inflammation, villus destruction, and enhanced enterocyte apoptosis in the intestines compared with CS-infected wild-type mice. CONCLUSIONS Our data suggest that depletion of PMNs and macrophages from the lamina propria (LP) exacerbates experimental NEC, indicating that both of these immunocytes play an important role in the clearance of CS during the initial stages of infection. The increased mucosal cytokine response and NO production in the absence of these immunocytes may be responsible for the observed increase in mucosal injury. Understanding how CS manipulates these cells, employing novel mouse model of NEC reported in this study, will provide significant insights for the development of novel therapeutic and preventive strategies to combat NEC.
Collapse
Affiliation(s)
- Claudia N Emami
- Department of Surgery, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
46
|
Bibliography. Neonatology and perinatology. Current world literature. Curr Opin Pediatr 2011; 23:253-7. [PMID: 21412083 DOI: 10.1097/mop.0b013e3283454167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
|
48
|
Khailova L, Mount Patrick SK, Arganbright KM, Halpern MD, Kinouchi T, Dvorak B. Bifidobacterium bifidum reduces apoptosis in the intestinal epithelium in necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2010; 299:G1118-27. [PMID: 20705904 PMCID: PMC2993641 DOI: 10.1152/ajpgi.00131.2010] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [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 a devastating intestinal disease of neonates, and clinical studies suggest the beneficial effect of probiotics in NEC prevention. Recently, we have shown that administration of Bifidobacterium bifidum protects against NEC in a rat model. Intestinal apoptosis can be suppressed by activation of cyclooxygenase-2 (COX-2) and increased production of prostaglandin E(2) (PGE(2)). The present study investigates the effect of B. bifidum on intestinal apoptosis in the rat NEC model and in an intestinal epithelial cell line (IEC-6), as a mechanism of protection against mucosal injury. Premature rats were divided into the following three groups: dam fed, hand fed with formula (NEC), or hand fed with formula supplemented with B. bifidum (NEC + B. bifidum). Intestinal Toll-like receptor-2 (TLR-2), COX-2, PGE(2), and apoptotic regulators were measured. The effect of B. bifidum was verified in IEC-6 cells using a model of cytokine-induced apoptosis. Administration of B. bifidum increased expression of TLR-2, COX-2, and PGE(2) and significantly reduced apoptosis in the intestinal epithelium of both in vivo and in vitro models. The Bax-to-Bcl-w ratio was shifted toward cell survival, and the number of cleaved caspase-3 positive cells was markedly decreased in B. bifidum-treated rats. Experiments in IEC-6 cells showed anti-apoptotic effect of B. bifidum. Inhibition of COX-2 signaling blocked the protective effect of B. bifidum treatment in both in vivo and in vitro models. In conclusion, oral administration of B. bifidum activates TLR-2 in the intestinal epithelium. B. bifidum increases expression of COX-2, which leads to higher production of PGE(2) in the ileum and protects against intestinal apoptosis associated with NEC. This study indicates the ability of B. bifidum to downregulate apoptosis in the rat NEC model and in IEC-6 cells by a COX-2-dependent matter and suggests a molecular mechanism by which this probiotic reduces mucosal injury and preserves intestinal integrity.
Collapse
Affiliation(s)
- Ludmila Khailova
- Department of 1Pediatrics and Steele Children's Research Center, and
| | | | | | | | - Toshi Kinouchi
- 2Food Science Institute, Meiji Dairies Corporation, Odawara, Japan
| | - Bohuslav Dvorak
- Department of 1Pediatrics and Steele Children's Research Center, and ,3Cell Biology & Anatomy, University of Arizona, Tucson, Arizona; and
| |
Collapse
|
49
|
Ellis CL, Rutledge JC, Underwood MA. Intestinal microbiota and blue baby syndrome: probiotic therapy for term neonates with cyanotic congenital heart disease. Gut Microbes 2010; 1:359-66. [PMID: 21468216 PMCID: PMC3056099 DOI: 10.4161/gmic.1.6.14077] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 09/15/2010] [Accepted: 10/29/2010] [Indexed: 02/03/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common intestinal emergency among premature infants. Risk factors in premature infants include immature intestinal immunity and an intestinal microbiota dominated by hospital-acquired bacteria. Some probiotics have been shown to decrease the incidence of NEC in premature infants. Among term infants, NEC is rare. However, among term infants with cyanotic congenital heart disease (CCHD), the incidence of NEC is similar to that of premature infants but with even greater mortality rates. Mechanisms by which NEC occurs in term infants with CCHD are unknown. Of central interest is the potential role of changes in the intestinal microbiota and whether these can be modified with probiotic bacteria; accordingly, we review the literature, propose hypotheses and present the rationale for future studies involving preliminary probiotic clinical trials.
Collapse
Affiliation(s)
- Collin L Ellis
- Department of Internal Medicine; Division of Cardiovascular Medicine; University of California at Davis; Davis, CA USA,Graduate Programs in Nutritional Biology, Biotechnology and Biophotonics; University of California at Davis; Davis, CA USA
| | - John C Rutledge
- Department of Internal Medicine; Division of Cardiovascular Medicine; University of California at Davis; Davis, CA USA,Graduate Programs in Nutritional Biology, Biotechnology and Biophotonics; University of California at Davis; Davis, CA USA
| | - Mark A Underwood
- Department of Pediatrics; Division of Neonatology; University of California at Davis; Davis, CA USA
| |
Collapse
|