51
|
Nasef M, Ben Turkia H, Haider Ali AM, Mahdawi E, Nair A. To What Extent Does Arginine Reduce the Risk of Developing Necrotizing Enterocolitis? Cureus 2023; 15:e45813. [PMID: 37876383 PMCID: PMC10591459 DOI: 10.7759/cureus.45813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Necrotizing enterocolitis (NEC) and neonatal sepsis are polar opposite diseases that are commonly encountered in the NICU. Concerning the frequency of these pathologies, NEC is regarded as being a much rarer condition, whereas neonatal sepsis is slightly more commonly encountered. However, neonatal sepsis can present with varying clinical presentations and, if caught late, can be detrimental to the patient. Many different modes of therapies have been studied for both conditions at different levels of pathologies, from a microscopic to a macroscopic level, leading to an assessment of treatment approaches. With the different ongoing treatment protocols being studied, one such therapy under investigation that does stand out is the use of L-arginine in both conditions. The L-arginine, being an essential amino acid, has many basic biological roles in developing neonates. It mainly involves the production of nitric oxide (NO), a potent vasodilator, which is particularly important in the development of vasculature in almost every organ. In premature infants, poorly developed vasculature makes them more susceptible to injury, therefore increasing the risk of diseases such as NEC and the severity of diseases such as neonatal sepsis. By assessing the uses of L-arginine and its application towards treating conditions like NEC and neonatal sepsis, we aim to identify its potential benefits as a treatment and its potential applications in clinical practice by understanding its basic functions and role in the pathophysiology of NEC and neonatal sepsis.
Collapse
Affiliation(s)
- Minoosh Nasef
- Neonatology, King Hamad University Hospital, Muharraq, BHR
| | | | | | - Esam Mahdawi
- Obstetrics and Gynaecology, King Hamad University Hospital, Muharraq, BHR
| | - Arun Nair
- Pediatrics, Saint Peter's University Hospital, Somerset, USA
| |
Collapse
|
52
|
Yeramilli V, Cheddadi R, Benjamin H, Martin C. The Impact of Stress, Microbial Dysbiosis, and Inflammation on Necrotizing Enterocolitis. Microorganisms 2023; 11:2206. [PMID: 37764050 PMCID: PMC10534571 DOI: 10.3390/microorganisms11092206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of intestinal morbidity and mortality in neonates. A large body of work exists; however, the pathogenesis of NEC remains poorly understood. Numerous predictors have been implicated in the development of NEC, with relatively less emphasis on maternal factors. Utilizing human tissue plays a crucial role in enhancing our comprehension of the underlying mechanisms accountable for this devastating disease. In this review, we will discuss how maternal stress affects the pathogenesis of NEC and how changes in the intestinal microbiome can influence the development of NEC. We will also discuss the results of transcriptomics-based studies and analyze the gene expression changes in NEC tissues and other molecular targets associated with the pathogenesis of NEC.
Collapse
Affiliation(s)
| | | | | | - Colin Martin
- Division of Pediatric, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. S., Lowder Building Suite 300, Birmingham, AL 35233, USA
| |
Collapse
|
53
|
Chen G, Lv X, Tang W. Fecal calprotectin as a non-invasive marker for the prediction of post-necrotizing enterocolitis stricture. Pediatr Surg Int 2023; 39:250. [PMID: 37594554 DOI: 10.1007/s00383-023-05534-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
PURPOSE This study aimed to evaluate the clinical utility of fecal calprotectin (FC) levels during the necrotizing enterocolitis (NEC) episode to predict the onset of post-NEC intestinal stricture. METHODS The medical records of patients with NEC treated from April 2020 to April 2022 were recorded for this study. FC was quantified at the acute phase of NEC. FC levels were compared in patients with or without intestinal stricture. Receiver operating characteristics (ROC) analysis was constructed to determine optimal cut-offs of FC for post-NEC intestinal stricture. RESULTS A total of 50 infants with NEC were enrolled in this study and 14 (28%) of them eventually developed intestinal stricture. All children with intestinal stricture underwent one-stage surgery and all made it through the follow-up period alive. The median FC level was 1237.55 (741.25, 1378.80) ug/g in patients with intestinal stricture and it was significantly higher than that in the non-stricture group [158.30 (76.23, 349.13) ug/g, P < 0.001]. FC had good diagnostic accuracy for predicting intestinal stricture, according to ROC curve analysis, with an AUC area of 0.911. At an optimal cut-off value of 664.2 ug/g, sensitivity and specificity were 85.71% and 91.67%, respectively. CONCLUSION As a non-invasive parameter, FC has excellent efficacy and accuracy in predicting post-NEC intestinal stricture. Increased FC levels at the acute phase of NEC were associated with the development of intestinal stricture.
Collapse
Affiliation(s)
- Guanglin Chen
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xiaofeng Lv
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
54
|
Luo Z, Chen A, Xie A, Liu X, Jiang S, Yu R. Limosilactobacillus reuteri in immunomodulation: molecular mechanisms and potential applications. Front Immunol 2023; 14:1228754. [PMID: 37638038 PMCID: PMC10450031 DOI: 10.3389/fimmu.2023.1228754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Frequent use of hormones and drugs may be associated with side-effects. Recent studies have shown that probiotics have effects on the prevention and treatment of immune-related diseases. Limosilactobacillus reuteri (L. reuteri) had regulatory effects on intestinal microbiota, host epithelial cells, immune cells, cytokines, antibodies (Ab), toll-like receptors (TLRs), tryptophan (Try) metabolism, antioxidant enzymes, and expression of related genes, and exhibits antibacterial and anti-inflammatory effects, leading to alleviation of disease symptoms. Although the specific composition of the cell-free supernatant (CFS) of L. reuteri has not been clarified, its efficacy in animal models has drawn increased attention to its potential use. This review summarizes the effects of L. reuteri on intestinal flora and immune regulation, and discusses the feasibility of its application in atopic dermatitis (AD), asthma, necrotizing enterocolitis (NEC), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS), and provides insights for the prevention and treatment of immune-related diseases.
Collapse
Affiliation(s)
- Zichen Luo
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ailing Chen
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Anni Xie
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Xueying Liu
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Renqiang Yu
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
55
|
Johnson-Hence CB, Gopalakrishna KP, Bodkin D, Coffey KE, Burr AH, Rahman S, Rai AT, Abbott DA, Sosa YA, Tometich JT, Das J, Hand TW. Stability and heterogeneity in the antimicrobiota reactivity of human milk-derived immunoglobulin A. J Exp Med 2023; 220:e20220839. [PMID: 37462916 PMCID: PMC10354535 DOI: 10.1084/jem.20220839] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 04/11/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Immunoglobulin A (IgA) is secreted into breast milk and is critical for both protecting against enteric pathogens and shaping the infant intestinal microbiota. The efficacy of breast milk-derived maternal IgA (BrmIgA) is dependent upon its specificity; however, heterogeneity in BrmIgA binding ability to the infant microbiota is not known. Using a flow cytometric array, we analyzed the reactivity of BrmIgA against bacteria common to the infant microbiota and discovered substantial heterogeneity between all donors, independent of preterm or term delivery. Surprisingly, we also observed intradonor variability in the BrmIgA response to closely related bacterial isolates. Conversely, longitudinal analysis showed that the antibacterial BrmIgA reactivity was relatively stable through time, even between sequential infants, indicating that mammary gland IgA responses are durable. Together, our study demonstrates that the antibacterial BrmIgA reactivity displays interindividual heterogeneity but intraindividual stability. These findings have important implications for how breast milk shapes the development of the preterm infant microbiota and protects against necrotizing enterocolitis.
Collapse
Affiliation(s)
- Chelseá B. Johnson-Hence
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kathyayini P. Gopalakrishna
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darren Bodkin
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara E. Coffey
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ansen H.P. Burr
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Syed Rahman
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ali T. Rai
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darryl A. Abbott
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yelissa A. Sosa
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Justin T. Tometich
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy W. Hand
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
56
|
Martins RDS, Kooi EMW, Poelstra K, Hulscher JBF. The role of intestinal alkaline phosphatase in the development of necrotizing enterocolitis. Early Hum Dev 2023; 183:105797. [PMID: 37300991 DOI: 10.1016/j.earlhumdev.2023.105797] [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: 04/28/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal disease that affects neonates worldwide and often leads to high morbidity and mortality rates. Despite extensive research, the cause of NEC remains unclear, and current treatment options are limited. An important novel finding is the potential role of intestinal Alkaline Phosphatase (IAP) in both pathogenesis and treatment of NEC. IAP can play a vital role in detoxifying liposaccharides (LPS), a key mediator of many pathological processes, thereby reducing the inflammatory response associated with NEC. Furthermore, IAP can help prevent dysbiosis, improve intestinal perfusion, and promote autophagy. In this comprehensive review, we present evidence of the possible connection between IAP and the LPS/Toll-like receptor 4 (TLR4) pathway, impaired gut immunity, and dysbiosis in the preterm gut. Based on these findings, the administration of exogenous IAP might provide promising preventive and therapeutic avenues for the management of NEC.
Collapse
Affiliation(s)
- Raquel Dos Santos Martins
- Division of Pediatric Surgery, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Elisabeth M W Kooi
- Division of Neonatology, Department of Pediatrics, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Klaas Poelstra
- Department of Nanomedicine and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, the Netherlands
| | - Jan B F Hulscher
- Division of Pediatric Surgery, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
57
|
Sami AS, Frazer LC, Miller CM, Singh DK, Clodfelter LG, Orgel KA, Good M. The role of human milk nutrients in preventing necrotizing enterocolitis. Front Pediatr 2023; 11:1188050. [PMID: 37334221 PMCID: PMC10272619 DOI: 10.3389/fped.2023.1188050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an intestinal disease that primarily impacts preterm infants. The pathophysiology of NEC involves a complex interplay of factors that result in a deleterious immune response, injury to the intestinal mucosa, and in its most severe form, irreversible intestinal necrosis. Treatments for NEC remain limited, but one of the most effective preventative strategies for NEC is the provision of breast milk feeds. In this review, we discuss mechanisms by which bioactive nutrients in breast milk impact neonatal intestinal physiology and the development of NEC. We also review experimental models of NEC that have been used to study the role of breast milk components in disease pathophysiology. These models are necessary to accelerate mechanistic research and improve outcomes for neonates with NEC.
Collapse
Affiliation(s)
- Ahmad S. Sami
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lynda G. Clodfelter
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
58
|
Mackay S, Frazer LC, Bailey GK, Miller CM, Gong Q, Dewitt ON, Singh DK, Good M. Identification of serum biomarkers for necrotizing enterocolitis using aptamer-based proteomics. Front Pediatr 2023; 11:1184940. [PMID: 37325361 PMCID: PMC10264655 DOI: 10.3389/fped.2023.1184940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Necrotizing enterocolitis (NEC) is a potentially fatal intestinal disease primarily affecting preterm infants. Early diagnosis of neonates with NEC is crucial to improving outcomes; however, traditional diagnostic tools remain inadequate. Biomarkers represent an opportunity to improve the speed and accuracy of diagnosis, but they are not routinely used in clinical practice. Methods In this study, we utilized an aptamer-based proteomic discovery assay to identify new serum biomarkers of NEC. We compared levels of serum proteins in neonates with and without NEC and identified ten differentially expressed serum proteins between these groups. Results We detected two proteins, C-C motif chemokine ligand 16 (CCL16) and immunoglobulin heavy constant alpha 1 and 2 heterodimer (IGHA1 IGHA2), that were significantly increased during NEC and eight that were significantly decreased. Generation of receiver operating characteristic (ROC) curves revealed that alpha-fetoprotein (AUC = 0.926), glucagon (AUC = 0.860), and IGHA1 IGHA2 (AUC = 0.826) were the proteins that best differentiated patients with and without NEC. Discussion These findings indicate that further investigation into these serum proteins as a biomarker for NEC is warranted. In the future, laboratory tests incorporating these differentially expressed proteins may improve the ability of clinicians to diagnose infants with NEC rapidly and accurately.
Collapse
Affiliation(s)
- Stephen Mackay
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Grace K. Bailey
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Qingqing Gong
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Olivia N. Dewitt
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| |
Collapse
|
59
|
García-Santos JA, Nieto-Ruiz A, García-Ricobaraza M, Cerdó T, Campoy C. Impact of Probiotics on the Prevention and Treatment of Gastrointestinal Diseases in the Pediatric Population. Int J Mol Sci 2023; 24:9427. [PMID: 37298377 PMCID: PMC10253478 DOI: 10.3390/ijms24119427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Despite the high prevalence of gastrointestinal disorders (GIDs) in infants and children, especially those categorized as functional GIDs (FGIDs), insufficient knowledge about their pathophysiology has limited both symptomatic diagnosis and the development of optimal therapies. Recent advances in the field of probiotics have made their potential use as an interesting therapeutic and preventive strategy against these disorders possible, but further efforts are still needed. In fact, there is great controversy surrounding this topic, generated by the high variety of potential probiotics strains with plausible therapeutic utility, the lack of consensus in their use as well as the few comparative studies available on probiotics that record their efficacy. Taking into account these limitations, and in the absence of clear guidelines about the dose and timeframe for successful probiotic therapy, our review aimed to evaluate current studies on potential use of probiotics for the prevention and treatment of the most common FGIDs and GIDs in the pediatric population. Furthermore, matters referring to know major action pathways and key safety recommendations for probiotic administration proposed by major pediatric health agencies shall also be discussed.
Collapse
Affiliation(s)
- José Antonio García-Santos
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (J.A.G.-S.); (A.N.-R.); (M.G.-R.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Avda del Conocimiento 19, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs-GRANADA), Health Sciences Technological Park, Avda. de Madrid 15, 18012 Granada, Spain
| | - Ana Nieto-Ruiz
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (J.A.G.-S.); (A.N.-R.); (M.G.-R.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Avda del Conocimiento 19, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs-GRANADA), Health Sciences Technological Park, Avda. de Madrid 15, 18012 Granada, Spain
| | - María García-Ricobaraza
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (J.A.G.-S.); (A.N.-R.); (M.G.-R.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Avda del Conocimiento 19, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs-GRANADA), Health Sciences Technological Park, Avda. de Madrid 15, 18012 Granada, Spain
| | - Tomás Cerdó
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (J.A.G.-S.); (A.N.-R.); (M.G.-R.)
- Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), Av. Menéndez Pidal, s/n, 14004 Córdoba, Spain
- Centre for Rheumatology Research, Division of Medicine, University College London, Gower Street, London WC1E 6BT, UK
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (J.A.G.-S.); (A.N.-R.); (M.G.-R.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Avda del Conocimiento 19, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs-GRANADA), Health Sciences Technological Park, Avda. de Madrid 15, 18012 Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada’s Node, Carlos III Health Institute, Avda. Monforte de Lemos 5, 28028 Madrid, Spain
| |
Collapse
|
60
|
Ganji N, Li B, Lee C, Pierro A. Necrotizing enterocolitis: recent advances in treatment with translational potential. Pediatr Surg Int 2023; 39:205. [PMID: 37247104 DOI: 10.1007/s00383-023-05476-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/30/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most prevalent and devastating gastrointestinal disorders in neonates. Despite advances in neonatal care, the incidence and mortality due to NEC remain high, highlighting the need to devise novel treatments for this disease. There have been a number of recent advancements in therapeutic approaches for the treatment of NEC; these involve remote ischemic conditioning (RIC), stem cell therapy, breast milk components (human milk oligosaccharides, exosomes, lactoferrin), fecal microbiota transplantation, and immunotherapy. This review summarizes the most recent advances in NEC treatment currently underway as well as their applicability and associated challenges and limitations, with the aim to provide new insight into the paradigm of care for NEC worldwide.
Collapse
Affiliation(s)
- Niloofar Ganji
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - Bo Li
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - Carol Lee
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - Agostino Pierro
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
61
|
Yang J, Shi Y. Paneth cell development in the neonatal gut: pathway regulation, development, and relevance to necrotizing enterocolitis. Front Cell Dev Biol 2023; 11:1184159. [PMID: 37266449 PMCID: PMC10231676 DOI: 10.3389/fcell.2023.1184159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Paneth cells (PCs) are intestinal epithelial cells (IECs) that contain eosinophilic granules, which are located in Lieberkühn crypts. An increasing number of animal and human experiments have indicated that PCs are involved in the progression of a variety of intestinal as well as systemic inflammatory responses including necrotizing enterocolitis (NEC). NEC is an enteric acquired disease with high mortality that usually occurs in premature infants and neonates, however the underlying mechanisms remain unclear. In this review, we summarize the features of PCs, including their immune function, association with gut microbiota and intestinal stem cells, and their mechanism of regulating IEC death to explore the possible mechanisms by which PCs affect NEC.
Collapse
|
62
|
Xiong X, Wang Y, Chen X, Lin B, Zhuang Y, Luo L, Wang H, Yang C. Mixed Feedings and Necrotizing Enterocolitis: The Proportion of Human Milk Matters. Breastfeed Med 2023. [PMID: 37184535 DOI: 10.1089/bfm.2022.0268] [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] [Indexed: 05/16/2023]
Abstract
Objectives: Impact of the proportion of human milk (HM) in mixed feeding on necrotizing enterocolitis (NEC) remains unknown. This study explores the influence of different proportions of HM on the risk of NEC. Materials and Methods: A retrospective cohort study was performed in infants with very low birth weight (VLBW). A spline smoothing curve was used to evaluate the dose-dependent association between HM and the risk of NEC. Univariate and multivariate analyses were performed to detect the association between the proportion of HM and NEC. Results: Twenty-four infants developed NEC, with 4 (1.9%) in the high HM group, 18 (28.1%) in the low HM group, and 2 (8.0%) in the exclusive formula group (p < 0.001). After adjusting for the relevant confounders, low HM (proportion of HM ≤54%) (OR 33.526, 95% confidential interval [CI] 7.183-156.475, p < 0.001) and exclusive formula feeding (OR 8.493, 95% CI 1.107-65.187, p = 0.040) significantly increased the incidence of NEC, compared with the high HM feeding (proportion of HM >54%). Similarly, low HM was independently associated with an increased risk of feeding intolerance compared with high HM feeding (OR 4.383, 95% CI 2.243-8.564, p < 0.001). Conclusion: A low ratio of HM (≤54%) significantly increased the risk of intestinal complications in VLBW infants. Mixed feeding should relate to the proportion of HM in premature infants.
Collapse
Affiliation(s)
- Xiaoyun Xiong
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
- School of Nursing, Philippine Women's University, Manila, Philippines
| | - Yanqiong Wang
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Xueyu Chen
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Bingchun Lin
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yanzhu Zhuang
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Li Luo
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Huiyan Wang
- Shanxi University of Chinese Medicine, College of nursing, Jinzhong, China
| | - Chuanzhong Yang
- Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| |
Collapse
|
63
|
Lanik WE, Luke CJ, Nolan LS, Gong Q, Frazer LC, Rimer JM, Gale SE, Luc R, Bidani SS, Sibbald CA, Lewis AN, Mihi B, Agrawal P, Goree M, Maestas M, Hu E, Peters DG, Good M. Microfluidic device facilitates in vitro modeling of human neonatal necrotizing enterocolitis-on-a-chip. JCI Insight 2023; 8:e146496. [PMID: 36881475 PMCID: PMC10243823 DOI: 10.1172/jci.insight.146496] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a deadly gastrointestinal disease of premature infants that is associated with an exaggerated inflammatory response, dysbiosis of the gut microbiome, decreased epithelial cell proliferation, and gut barrier disruption. We describe an in vitro model of the human neonatal small intestinal epithelium (Neonatal-Intestine-on-a-Chip) that mimics key features of intestinal physiology. This model utilizes intestinal enteroids grown from surgically harvested intestinal tissue from premature infants and cocultured with human intestinal microvascular endothelial cells within a microfluidic device. We used our Neonatal-Intestine-on-a-Chip to recapitulate NEC pathophysiology by adding infant-derived microbiota. This model, named NEC-on-a-Chip, simulates the predominant features of NEC, including significant upregulation of proinflammatory cytokines, decreased intestinal epithelial cell markers, reduced epithelial proliferation, and disrupted epithelial barrier integrity. NEC-on-a-Chip provides an improved preclinical model of NEC that facilitates comprehensive analysis of the pathophysiology of NEC using precious clinical samples. This model is an advance toward a personalized medicine approach to test new therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Wyatt E. Lanik
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Cliff J. Luke
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lila S. Nolan
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qingqing Gong
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lauren C. Frazer
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jamie M. Rimer
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sarah E. Gale
- Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Shay S. Bidani
- Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Angela N. Lewis
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Belgacem Mihi
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pranjal Agrawal
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Martin Goree
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marlie Maestas
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elise Hu
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - David G. Peters
- University of Pittsburgh and Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Misty Good
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
64
|
Shi B, Shen L, Huang W, Cai L, Yang S, Zhang Y, Tou J, Lai D. A Nomogram for Predicting Surgical Timing in Neonates with Necrotizing Enterocolitis. J Clin Med 2023; 12:jcm12093062. [PMID: 37176503 PMCID: PMC10179100 DOI: 10.3390/jcm12093062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/02/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
OBJECTIVE To explore the surgical risk variables in patients with necrotizing enterocolitis (NEC) and develop a nomogram model for predicting the surgical intervention timing of NEC. METHODS Infants diagnosed with NEC were enrolled in our study. We gathered information from clinical data, laboratory examinations, and radiological manifestations. Using LASSO (least absolute shrinkage and selection operator) regression analysis and multivariate logistic regression analysis, a clinical prediction model based on the logistic nomogram was developed. The performance of the nomogram model was evaluated using the receiver operating characteristic (ROC) curve, calibration curves, and decision curve analysis (DCA). RESULTS A surgical intervention risk nomogram based on hypothermia, absent bowel sounds, WBC > 20 × 109/L or < 5 × 109/L, CRP > 50 mg/L, pneumatosis intestinalis, and ascites was practical, had a moderate predictive value (AUC > 0.8), improved calibration, and enhanced clinical benefit. CONCLUSIONS This simple and reliable clinical prediction nomogram model can help physicians evaluate children with NEC in a fast and effective manner, enabling the early identification and diagnosis of children at risk for surgery. It offers clinical revolutionary value for the development of medical or surgical treatment plans for children with NEC.
Collapse
Affiliation(s)
- Bo Shi
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Leiting Shen
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Wenchang Huang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Linghao Cai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Yuanyuan Zhang
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Binjiang Institute of Zhejiang University, Hangzhou 310053, China
| |
Collapse
|
65
|
Xia J, Claud EC. Gut Microbiome-Brain Axis as an Explanation for the Risk of Poor Neurodevelopment Outcome in Preterm Infants with Necrotizing Enterocolitis. Microorganisms 2023; 11:1035. [PMID: 37110458 PMCID: PMC10142133 DOI: 10.3390/microorganisms11041035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Necrotizing Enterocolitis (NEC) is characterized by an inflammation of intestinal tissue that primarily affects premature infants. It is the most common and devastating gastrointestinal morbidity of prematurity, but beyond intestinal morbidity, this condition has also been associated with an increased risk of neurodevelopmental delays that persist beyond infancy. Prematurity, enteral feeding, bacterial colonization, and prolonged exposure to antibiotics are all risk factors that predispose preterm infants to NEC. Interestingly, these factors are all also associated with the gut microbiome. However, whether or not there is a connection between the microbiome and the risk of neurodevelopmental delays in infants after NEC is still an emerging area of research. Furthermore, how microbes in the gut could impact a distant organ such as the brain is also poorly understood. In this review, we discuss the current understanding of NEC and the role of the gut microbiome-brain axis in neurodevelopmental outcomes after NEC. Understanding the potential role of the microbiome in neurodevelopmental outcomes is important as the microbiome is modifiable and thus offers the hope of improved therapeutic options. We highlight the progress and limitations in this field. Insights into the gut microbiome-brain axis may offer potential therapeutic approaches to improve the long-term outcomes of premature infants.
Collapse
Affiliation(s)
- Jason Xia
- College of Liberal Arts and Sciences, University of Illinois Urbana-Champion, Champaign, IL 61801, USA
| | - Erika C. Claud
- Department of Pediatrics and Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
66
|
Hoffsten A, Lilja HE, Mobini-Far H, Sindelar R, Markasz L. Paneth cell proteins DEFA6 and GUCA2A as tissue markers in necrotizing enterocolitis. Eur J Pediatr 2023:10.1007/s00431-023-04907-3. [PMID: 37017768 PMCID: PMC10257617 DOI: 10.1007/s00431-023-04907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 04/06/2023]
Abstract
Previous studies suggest that Paneth cells are involved in NEC development. Defensin alpha 6 (DEFA6) and guanylate cyclase activator 2A (GUCA2A) are selective protein markers of Paneth cells. The objective was to explore DEFA6 and GUCA2A expression in intestinal tissue samples from newborn infants with and without NEC. Tissue samples from histologically intact intestine were analyzed from 70 infants: 43 underwent bowel resection due to NEC and 27 controls were operated due to conditions such as intestinal atresia, dysmotility, aganglionosis, pseudo-obstruction or volvulus. Each tissue sample was immunohistochemically stained for DEFA6 and GUCA2A. Semi-automated digital image analysis was performed to determine protein expression. Clinical data and protein expressions were compared between the groups. DEFA6 expression was lower in the NEC group (p = 0.006). Low DEFA6 correlated with risk of developing NEC in a logistic regression analysis, independently of gestational age and birth weight (OR 0.843 [CI 0.732-0.971]; p = 0.018). GUCA2A expression did not differ between the two groups. CONCLUSION Lower expression of DEFA6 together with intact GUCA2A expression indicates that NEC patients have well-defined Paneth cells but diminished defensin activity. Our results suggest that DEFA6 could be used as a biomarker for NEC. WHAT IS KNOWN • Previous studies of defensin activity in NEC have been inconsistent, showing that defensin levels may be increased or diminished in NEC. GUCA2A has to our knowledge never been studied in NEC. WHAT IS NEW • This study benchmarks two specific Paneth cell markers (DEFA6 and GUCA2A) and their activity in individuals with and without NEC. • The key finding is that the NEC group had a lower DEFA6 expression compared to the Controls, while the expression of GUCA2A did not differ between the groups.
Collapse
Affiliation(s)
- Alice Hoffsten
- Department of Women's and Children's Health, Uppsala University, Uppsala, SE-751 85, Sweden.
| | - Helene Engstrand Lilja
- Department of Women's and Children's Health, Uppsala University, Uppsala, SE-751 85, Sweden
- Section of Pediatric Surgery, University Children's Hospital, Uppsala, Sweden
| | - Hamid Mobini-Far
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Richard Sindelar
- Department of Women's and Children's Health, Uppsala University, Uppsala, SE-751 85, Sweden
- Neonatal Intensive Care Unit, University Children's Hospital, Uppsala, Sweden
| | - Laszlo Markasz
- Department of Women's and Children's Health, Uppsala University, Uppsala, SE-751 85, Sweden
- Neonatal Intensive Care Unit, University Children's Hospital, Uppsala, Sweden
| |
Collapse
|
67
|
Bautista GM, Cera AJ, Chaaban H, McElroy SJ. State-of-the-art review and update of in vivo models of necrotizing enterocolitis. Front Pediatr 2023; 11:1161342. [PMID: 37082706 PMCID: PMC10112335 DOI: 10.3389/fped.2023.1161342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/13/2023] [Indexed: 04/22/2023] Open
Abstract
NEC remains one of the most common causes of mortality and morbidity in preterm infants. Animal models of necrotizing enterocolitis (NEC) have been crucial in improving our understanding of this devastating disease and identifying biochemical pathways with therapeutic potential. The pathogenesis of NEC remains incompletely understood, with no specific entity that unifies all infants that develop NEC. Therefore, investigators rely on animal models to manipulate variables and provide a means to test interventions, making them valuable tools to enhance our understanding and prevent and treat NEC. The advancements in molecular analytic tools, genetic manipulation, and imaging modalities and the emergence of scientific collaborations have given rise to unique perspectives and disease correlates, creating novel pathways of investigation. A critical review and understanding of the current phenotypic considerations of the highly relevant animal models of NEC are crucial to developing novel therapeutic and preventative strategies for NEC.
Collapse
Affiliation(s)
- Geoanna M. Bautista
- Department of Pediatrics, Division of Neonatology, University of California, Davis, Sacramento, CA, United States
| | - Anjali J. Cera
- Department of Pediatrics, Division of Neonatology, University of California, Davis, Sacramento, CA, United States
| | - Hala Chaaban
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Steven J. McElroy
- Department of Pediatrics, Division of Neonatology, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
68
|
Everhart KC, Iskersky VN, Donevant SB, Wirth MD, Dail RB. Pathogenesis of Necrotizing Enterocolitis in Preterm Infants: An Adaptation of Neuman's Systems Theory. Nurs Sci Q 2023; 36:152-157. [PMID: 36994958 DOI: 10.1177/08943184221150264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Preterm infants have increased morbidity and mortality due to immature physiology and neonatal complications. Necrotizing enterocolitis (NEC) is a devastating gastrointestinal complication that affects morbidity and mortality in preterm infants. The authors present an adaptation of Neuman's systems model called the NEC systems model to explore intrinsic and extrinsic factors leading to NEC in preterm infants. The literature was searched for theoretical models to guide exploration of neonatal disease influenced by the environment. Neuman's Systems Model provides a holistic approach to care of the system, offering a foundation to develop frameworks to examine preterm infants in their environment with associated stressors.
Collapse
Affiliation(s)
- Kayla C Everhart
- Doctoral Student and Research Coordinator, University of South Carolina-College of Nursing, Columbia, South Carolina, USA
| | - Victor N Iskersky
- Director of Neonatology Prisma Health Midlands, Medical Director Prisma Health Richland, Neonatal Intensive Care Unit, Clinical Associate Professor of Pediatrics, School of Medicine University of South Carolina, Columbia, South Carolina, USA
| | - Sara B Donevant
- Assistant Professor, University of South Carolina-College of Nursing, Columbia, South Carolina, USA
| | - Michael D Wirth
- Assistant Professor, College of Nursing and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Robin B Dail
- Associate Dean for Faculty Affairs and Professor, University of South Carolina-College of Nursing, Columbia, South Carolina, USA
| |
Collapse
|
69
|
Bednarczyk MM, Żelazna O, Głuszczak‐Idziakowska E, Schreiber‐Zamora J, Furmanek M, Kociszewska‐Najman B. A case of necrotizing enterocolitis in full-term infant on his first day of life-Early presentation with multifactorial etiology. Clin Case Rep 2023; 11:e7150. [PMID: 37096169 PMCID: PMC10122096 DOI: 10.1002/ccr3.7150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
We aimed to show the complexity of NEC in the full-term newborns. Additionally, we wanted to underline the importance of breastfeeding from the first day of life and show how deteriorating effect the antibiotics can have on gastrointestinal tract.
Collapse
Affiliation(s)
| | - Olga Żelazna
- Student Scientific Group at the Department Neonatology “Proneo”Medical University of WarsawWarsawPoland
| | | | | | - Mariusz Furmanek
- Department of Pediatric RadiologyMedical University of WarsawWarsawPoland
| | | |
Collapse
|
70
|
Magner C, Jenkins D, Koc F, Tan MH, O'Toole M, Boyle J, Maguire N, Duignan S, Murphy K, Ross P, Stanton C, McMahon CJ. Protocol for a prospective cohort study exploring the gut microbiota of infants with congenital heart disease undergoing cardiopulmonary bypass (the GuMiBear study). BMJ Open 2023; 13:e067016. [PMID: 37001916 PMCID: PMC10069492 DOI: 10.1136/bmjopen-2022-067016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023] Open
Abstract
INTRODUCTION The gut microbiota develops from birth and matures significantly during the first 24 months of life, playing a major role in infant health and development. The composition of the gut microbiota is influenced by several factors including mode of delivery, gestational age, feed type and treatment with antibiotics. Alterations in the pattern of gut microbiota development and composition can be associated with illness and compromised health outcomes.Infants diagnosed with 'congenital heart disease' (CHD) often require surgery involving cardiopulmonary bypass (CPB) early in life. The impact of this type of surgery on the integrity of the gut microbiome is poorly understood. In addition, these infants are at significant risk of developing the potentially devastating intestinal condition necrotising enterocolitis. METHODS AND ANALYSIS This study will employ a prospective cohort study methodology to investigate the gut microbiota and urine metabolome of infants with CHD undergoing surgery involving CPB. Stool and urine samples, demographic and clinical data will be collected from eligible infants based at the National Centre for Paediatric Cardiac Surgery in Ireland. Shotgun metagenome sequencing will be performed on stool samples and urine metabolomic analysis will identify metabolic biomarkers. The impact of the underlying diagnosis, surgery involving CPB, and the influence of environmental factors will be explored. Data from healthy age-matched infants from the INFANTMET study will serve as a control for this study. ETHICS AND DISSEMINATION This study has received full ethical approval from the Clinical Research Ethics Committee of Children's Health Ireland, GEN/826/20.
Collapse
Affiliation(s)
- Claire Magner
- School of Nursing, Midwifery and Health Systems, University College Dublin, Dublin, Ireland
| | - Dominic Jenkins
- Laboratory, Children's Health Ireland at Crumlin, Crumlin, Ireland
| | - Fatma Koc
- School of Microbiology, University College Cork, Cork, Ireland
- Food Biosciences, Teagasc Food Research Centre, Cork, Ireland
| | - Mong Hoi Tan
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Molly O'Toole
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Jordan Boyle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Niamh Maguire
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Sophie Duignan
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Kiera Murphy
- University College Cork APC Microbiome Institute, Cork, Ireland
- Food Biosciences, Teagasc Food Research Centre, Moorepark, Ireland
| | - Paul Ross
- University College Cork College of Science Engineering and Food Science, Cork, Ireland
| | - Catherine Stanton
- University College Cork APC Microbiome Institute, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
| | - Colin J McMahon
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
- University College Dublin School of Medicine, Dublin, Ireland
| |
Collapse
|
71
|
Johnson-Hence CB, Gopalakrishna KP, Bodkin D, Coffey KE, Burr AH, Rahman S, Rai AT, Abbott DA, Sosa YA, Tometich JT, Das J, Hand TW. Stability and heterogeneity in the anti-microbiota reactivity of human milk-derived Immunoglobulin A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532940. [PMID: 36993366 PMCID: PMC10055037 DOI: 10.1101/2023.03.16.532940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
UNLABELLED Immunoglobulin A (IgA) is secreted into breast milk and is critical to both protecting against enteric pathogens and shaping the infant intestinal microbiota. The efficacy of breast milk-derived maternal IgA (BrmIgA) is dependent upon its specificity, however heterogeneity in BrmIgA binding ability to the infant microbiota is not known. Using a flow cytometric array, we analyzed the reactivity of BrmIgA against bacteria common to the infant microbiota and discovered substantial heterogeneity between all donors, independent of preterm or term delivery. We also observed intra-donor variability in the BrmIgA response to closely related bacterial isolates. Conversely, longitudinal analysis showed that the anti-bacterial BrmIgA reactivity was relatively stable through time, even between sequential infants, indicating that mammary gland IgA responses are durable. Together, our study demonstrates that the anti-bacterial BrmIgA reactivity displays inter-individual heterogeneity but intra-individual stability. These findings have important implications for how breast milk shapes the development of the infant microbiota and protects against Necrotizing Enterocolitis. SUMMARY We analyze the ability of breast milk-derived Immunoglobulin A (IgA) antibodies to bind the infant intestinal microbiota. We discover that each mother secretes into their breast milk a distinct set of IgA antibodies that are stably maintained over time.
Collapse
Affiliation(s)
- Chelseá B. Johnson-Hence
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center
| | - Kathyayini P. Gopalakrishna
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Darren Bodkin
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Kara E. Coffey
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Pediatrics, Division of Allergy and Immunology, University of Pittsburgh School of Medicine
| | - Ansen H.P. Burr
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Immunology, University of Pittsburgh School of Medicine
| | - Syed Rahman
- Department of Immunology, University of Pittsburgh School of Medicine
- Center for Systems Immunology, University of Pittsburgh School of Medicine
| | - Ali T. Rai
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Darryl A. Abbott
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Yelissa A. Sosa
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Justin T. Tometich
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh School of Medicine
- Center for Systems Immunology, University of Pittsburgh School of Medicine
| | - Timothy W. Hand
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Immunology, University of Pittsburgh School of Medicine
| |
Collapse
|
72
|
Wang P, Li T, Niu C, Sun S, Liu D. ROS-activated MAPK/ERK pathway regulates crosstalk between Nrf2 and Hif-1α to promote IL-17D expression protecting the intestinal epithelial barrier under hyperoxia. Int Immunopharmacol 2023; 116:109763. [PMID: 36736221 DOI: 10.1016/j.intimp.2023.109763] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 02/04/2023]
Abstract
Reactive oxygen species (ROS) damage to the intestinal barrier is a side effect of prolonged hyperoxia therapy in neonates, which impairs growth and development of the intestine and promotes intestinal diseases. However, the research on clinical prevention and treatment is lacking. Therefore, we investigated the molecular mechanisms of the neonate intestinal response against hyperoxia-derived ROS to find targets for intestinal barrier damage prevention. Human intestinal epithelial cells were incubated under hyperoxia (85% oxygen) to build an in vitro model. ROS and the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway were inhibited to detect the MAPK/ERK pathway, nuclear factor erythroid factor 2-related factor 2 (Nrf2), hypoxia-inducible factor-1α (Hif-1α), and interleukin-17D (IL-17D) expression. Nrf2 was inhibited to detect Hif-1α and IL-17D expression. Hif-1α was inhibited to detect Nrf2, IL-17D, and tight junction proteins expression and apoptosis. Cells were treated with human recombinant IL-17D to detect TNF-α, IL-1β, IL-10, and tight junction proteins expression. ROS, Nrf2, Hif-1α, and IL-17D were upregulated and the MAPK/ERK pathway was activated under hyperoxia. But ROS inhibition downregulated the MAPK/ERK pathway, Nrf2, Hif-1α, and IL-17D. MAPK/ERK pathway inhibition downregulated Nrf2, Hif-1α, and IL-17D. Nrf2 inhibition downregulated Hif-1α and IL-17D. Hif-1α inhibition downregulated Nrf2, IL-17D, tight junction proteins, and exacerbated apoptosis. The recombinant IL-17D downregulated TNF-α, IL-1β, but upregulated IL-10 and tight junction proteins. We concluded that Hyperoxia-generated ROS activated the MAPK/ERK pathway to regulate Nrf2, Hif-1α, and IL-17D expression. Nrf2 and Hif-1α were interdependent and promoted IL-17D. Importantly, Hif-1α and IL-17D expression protected the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Pingchuan Wang
- ShengJing Hospital of China Medical University, Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, SanHao Street No.36, HePing District, ShenYang, Liaoning 110000, China
| | - Tianming Li
- ShengJing Hospital of China Medical University, Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, SanHao Street No.36, HePing District, ShenYang, Liaoning 110000, China
| | - Changping Niu
- ShengJing Hospital of China Medical University, Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, SanHao Street No.36, HePing District, ShenYang, Liaoning 110000, China
| | - Siyu Sun
- ShengJing Hospital of China Medical University, Department of Gastroenterology, SanHao Street No.36, HePing District, ShenYang, Liaoning 110000, China
| | - Dongyan Liu
- ShengJing Hospital of China Medical University, Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, SanHao Street No.36, HePing District, ShenYang, Liaoning 110000, China.
| |
Collapse
|
73
|
Fang D, Xu T, Sun J, Shi J, Li F, Yin Y, Wang Z, Liu Y. Nicotinamide Mononucleotide Ameliorates Sleep Deprivation-Induced Gut Microbiota Dysbiosis and Restores Colonization Resistance against Intestinal Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207170. [PMID: 36698264 PMCID: PMC10037695 DOI: 10.1002/advs.202207170] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Indexed: 06/12/2023]
Abstract
Gut microbiota-mediated colonization resistance (CR) is crucial in protecting the host from intestinal infections. Sleep deprivation (SD) is an important contributor in the disturbances of intestinal homeostasis. However, whether and how SD affects host CR remains largely unknown. Here, it is shown that SD impairs intestinal CR in mice, whereas nicotinamide mononucleotide (NMN) supplementation restores it. Microbial diversity and metabolomic analyses suggest that gut microbiota and metabolite profiles in SD-treated mice are highly shaped, whereas NMN reprograms these differences. Specifically, the altered gut microbiota in SD mice further incurs the disorder of secondary bile acids pool accompanied by a decrease in deoxycholic acid (DCA). Conversely, NMN supplementation retakes the potential benefits of DCA, which is associated with specific gut microbiota involved in primary bile acids metabolic flux. In animal models of infection, DCA is effective in preventing and treating bacterial infections when used alone or in combination with antibiotics. Mechanistically, DCA alone disrupts membrane permeability and aggravates oxidative damage, thereby reducing intestinal pathogen burden. Meanwhile, exogenous DCA promotes antibiotic accumulation and destroys oxidant-antioxidant system, thus potentiating antibiotic efficacy. Overall, this work highlights the important roles of gut microbiota and bile acid metabolism in the maintenance of intestinal CR.
Collapse
Affiliation(s)
- Dan Fang
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Tianqi Xu
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Jingyi Sun
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Jingru Shi
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Fulei Li
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Yanqing Yin
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Zhiqiang Wang
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesJoint International Research Laboratory of Agriculture and Agri‐Product Safety the Ministry of Education of ChinaYangzhou UniversityYangzhou225009P. R. China
- Institute of Comparative MedicineYangzhou UniversityYangzhou225009P. R. China
| | - Yuan Liu
- College of Veterinary MedicineYangzhou UniversityYangzhou225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesJoint International Research Laboratory of Agriculture and Agri‐Product Safety the Ministry of Education of ChinaYangzhou UniversityYangzhou225009P. R. China
- Institute of Comparative MedicineYangzhou UniversityYangzhou225009P. R. China
| |
Collapse
|
74
|
Wang H, Meng X, Xing S, Guo B, Chen Y, Pan YQ. Probiotics to prevent necrotizing enterocolitis and reduce mortality in neonates: A meta-analysis. Medicine (Baltimore) 2023; 102:e32932. [PMID: 36827026 PMCID: PMC11309691 DOI: 10.1097/md.0000000000032932] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Probiotics are gradually being used as a supplementation to prevent necrotizing enterocolitis (NEC) and reduce mortality in neonates. We performed an updated meta-analysis to systematically evaluate the efficacy and safety of prophylactic probiotic supplementation for preventing NEC. METHODS The databases including PubMed, Embase, Scopus, Web of Science, and China National Knowledge Infrastructure were used to search the relevant articles. The latest retrieval date was up to December 2021. The meta-analysis was performed using Stata version 10.0. Finally, a total of 70 studies containing 8319 cases and 9283 controls were included. The strength of the association between the supplementation of probiotics and NEC was measured by risk ratios (RRs) with 95% confidence intervals (CIs). Pooled effect sizes across studies were performed by a random effect model. RESULTS The results showed that the probiotics could significantly reduce the incidence of NEC (stage II or more) (RR = 0.436, 95% CI = 0.357-0.531, P < .001), the overall mortality (RR = 0.651, 95% CI = 0.506-0.836, P < .001), and NEC-related mortality (RR = 0.639, 95% CI = 0.423-0.966, P = .034). Due to the lack of sufficient sample size, we did not perform the subgroup analysis by types of probiotic strain. CONCLUSION This meta-analysis indicates that the use of probiotics can effectively reduce the occurrence of NEC and mortality in neonates.
Collapse
Affiliation(s)
- Hongbo Wang
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Xinyao Meng
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Shihan Xing
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Baotong Guo
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Yuhan Chen
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Yu-Qing Pan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|
75
|
Screening inflammatory protein biomarkers on premature infants with necrotizing enterocolitis. Inflamm Res 2023; 72:757-768. [PMID: 36806964 PMCID: PMC10129932 DOI: 10.1007/s00011-023-01702-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 02/04/2023] [Indexed: 02/21/2023] Open
Abstract
OBJECTIVE This study aimed to explore potential inflammatory biomarkers for early prediction of necrotizing enterocolitis (NEC) in premature infants. METHODS Plasma samples were collected from premature infants with NEC (n = 30), sepsis (n = 29), and controls without infection (n = 29). The 92 inflammatory-related proteins were assessed via high-throughput OLINK proteomics platform. RESULTS There were 11 inflammatory proteins that significate differences (p < 0.05) among NEC, sepsis and control preterm infants, which include IL-8, TRAIL, IL-24, MMP-10, CCL20, CXCL1, OPG, TSLP, MCP-4, TNFSF14 and LIF. A combination of these 11 proteins could serve as differential diagnosis between NEC and control infants (AUC = 0.972), or between NEC and sepsis infants (AUC = 0.881). Furthermore, the combination of IL-8, OPG, MCP-4, IL-24, LIF and CCL20 could distinguish Stage II and III of NEC (AUC = 0.977). Further analysis showed the combination of IL-8, IL-24 and CCL20 have the best prediction value for NEC and control (AUC = 0.947), NEC and sepsis (AUC = 0.838) and different severity of NEC (AUC = 0.842). CONCLUSION Inflammatory proteins were different expressed in premature infants with NEC compared with controls or sepsis. Combining these proteins provide a higher diagnostic potential for preterm NEC infants.
Collapse
|
76
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
77
|
余 增, 许 淑, 孙 慧, 李 明, 邢 珊, 程 萍, 张 宏, 王 颖, 杨 子. [Clinical characteristics and risk factors for early-onset necrotizing enterocolitis in preterm infants with very/extremely low birth weight]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:147-152. [PMID: 36854690 PMCID: PMC9979387 DOI: 10.7499/j.issn.1008-8830.2208099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/14/2022] [Indexed: 03/02/2023]
Abstract
OBJECTIVES To investigate the clinical characteristics and risk factors for early-onset necrotizing enterocolitis (NEC) in preterm infants with very/extremely low birth weight (VLBW/ELBW). METHODS A retrospective analysis was performed on the medical data of 194 VLBW/ELBW preterm infants with NEC who were admitted to Children's Hospital Affiliated to Zhengzhou University from January 2014 to December 2021. These infants were divided into early-onset group (onset in the first two weeks of life; n=62) and late-onset group (onset two weeks after birth; n=132) based on their onset time. The two groups were compared in terms of perinatal conditions, clinical characteristics, laboratory examination results, and clinical outcomes. Sixty-two non-NEC infants with similar gestational age and birth weight who were hospitalized at the same period as these NEC preterm infants were selected as the control group. The risk factors for the development of early-onset NEC were identified using multivariate logistic regression analysis. RESULTS Compared with the late-onset group, the early-onset group had significantly higher proportions of infants with 1-minute Apgar score ≤3, stage III NEC, surgical intervention, grade ≥3 intraventricular hemorrhage, apnea, and fever or hypothermia (P<0.05). The multivariate logistic regression analysis showed that feeding intolerance, blood culture-positive early-onset sepsis, severe anemia, and hemodynamically significant patent ductus arteriosus were independent risk factors for the development of early-onset NEC in VLBW/ELBW preterm infants (P<0.05). CONCLUSIONS VLBW/ELBW preterm infants with early-onset NEC have more severe conditions compared with those with late-onset NEC. Neonates with feeding intolerance, blood culture-positive early-onset sepsis, severe anemia, or hemodynamically significant patent ductus arteriosus have a higher risk of early-onset NEC.
Collapse
|
78
|
Microbiota-dependent presence of murine enteric glial cells requires myeloid differentiation primary response protein 88 signaling. J Biosci 2023. [DOI: 10.1007/s12038-023-00325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
79
|
Inarejos Clemente EJ, Barber I, Navallas Irujo M, Ladera E, Sousa P, Salas B, Fernández CV, Rodríguez-Fanjul J, Navarro OM. US for Evaluation of Acute Abdominal Conditions in Neonates. Radiographics 2023; 43:e220110. [PMID: 36602924 DOI: 10.1148/rg.220110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
US is the imaging modality of choice for evaluation of a variety of abdominal conditions, and in recent years it has also become useful and promising as a bedside technique for assessment of acute abdominal conditions in neonates. Bedside US can help, complement, and sometimes replace radiographic or contrast-enhanced studies in critically ill and labile neonates who are difficult to transport to the fluoroscopy suite. Some of the features of bedside US can be applied as point-of-care US (POCUS) of the sick neonate. Some of the abdominal conditions in neonates that can be assessed and monitored with bedside US are necrotizing enterocolitis and its complications, malrotation with a midgut volvulus, segmental volvulus, meconium peritonitis, and complicated inguinal hernia. High-resolution US with the use of 15-MHz and higher-frequency probes allows characterization of the bowel anatomy and features of intestinal abnormalities in neonates in fine detail. Color Doppler US and microvascular imaging improve accuracy in the detection and characterization of bowel vascularity, which is important in the treatment and follow-up of patients with intestinal conditions. © RSNA, 2023 Quiz questions for this article are available through the Online Learning Center. The slide presentation from the RSNA Annual Meeting is available for this article.
Collapse
Affiliation(s)
- Emilio J Inarejos Clemente
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Ignasi Barber
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - María Navallas Irujo
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Enrique Ladera
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Paulino Sousa
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Bárbara Salas
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Carmen Virginia Fernández
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Javier Rodríguez-Fanjul
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| | - Oscar M Navarro
- From the Department of Diagnostic Imaging (E.J.I.C., I.B., M.N.I., E.L., P.S., B.S.) and Neonatal Intensive Care Unit, Department of Pediatrics (C.V.F.), Hospital Sant Joan de Déu, Av Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona, Spain (J.R.F.); and Department of Medical Imaging, University of Toronto, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada (O.M.N.)
| |
Collapse
|
80
|
Zhang X, Zhang Y, He Y, Zhu X, Ai Q, Shi Y. β-glucan protects against necrotizing enterocolitis in mice by inhibiting intestinal inflammation, improving the gut barrier, and modulating gut microbiota. J Transl Med 2023; 21:14. [PMID: 36627673 PMCID: PMC9830848 DOI: 10.1186/s12967-022-03866-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease with high morbidity and mortality, affecting preterm infants especially those with very low and extremely low birth weight. β-glucan has manifested multiple biological effects including anti-inflammatory, regulation of gut microbiota, and immunomodulatory activities. This study aimed to investigate the effects of β-glucan on NEC. METHODS Neonatal C57BL/6 mice were randomly divided into three groups: Control group, NEC group and β-glucan group. Newborn 3-day-old mice were gavaged with either 1 mg/ml β-glucan or phosphate buffer saline at 0.03 ml/g for 7 consecutive days before NEC induction and a NEC model was established with hypoxia combined with cold exposure and formula feeding. All the pups were killed after 72-h modeling. Hematoxylin-eosin staining was performed to assess the pathological injury to the intestines. The mRNA expression levels of inflammatory factors in intestinal tissues were determined using quantitative real-time PCR. The protein levels of TLR4, NF-κB and tight junction proteins in intestinal tissues were evaluated using western blotting and immunohistochemistry. 16S rRNA sequencing was performed to determine the structure of the gut microbiota. RESULTS β-glucan administration ameliorated intestinal injury of NEC mice; reduced the intestinal expression of TLR4, NF-κB, IL-1β, IL-6, and TNF-α; increased the intestinal expression of IL-10; and improved the expression of ZO-1, Occludin and Claudin-1 within the intestinal barrier. Pre-treatment with β-glucan also increased the proportion of Actinobacteria, Clostridium butyricum, Lactobacillus johnsonii, Lactobacillus murinus, and Lachnospiraceae bacterium mt14 and reduced the proportion of Klebsiella oxytoca g Klebsiella in the NEC model. CONCLUSION β-glucan intervention prevents against NEC in neonatal mice, possibly by suppressing the TLR4-NF-κB signaling pathway, improving intestinal barrier function, and partially regulating intestinal microbiota.
Collapse
Affiliation(s)
- Xingdao Zhang
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuni Zhang
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xingwang Zhu
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qing Ai
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuan Shi
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
81
|
FERDINANDO DLT, FRAGA FDCB, PIATTO VB, SOUZA AS. IS -94INS/DELATTG POLYMORPHISM IN THE NUCLEAR FACTOR KAPPA-B1 GENE (NFKB1) ASSOCIATED WITH NECROTIZING ENTEROCOLITIS? ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2023; 35:e1717. [PMID: 36629694 PMCID: PMC9830673 DOI: 10.1590/0102-672020220002e1717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/19/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Abnormalities in the different stages of the intestinal maturation process cause metabolic and molecular changes. Among the genetic alterations associated with necrotizing enterocolitis, the -94ins/delATTG polymorphism in NFKB1 gene leads to unregulated activation of the NFKB protein due to an increase in the inherent pro-inflammatory state of the premature intestine. AIMS To determine the prevalence of the -94ins/delATTG polymorphism in NFKB1 gene in neonates with and without necrotizing enterocolitis. METHODS This is a case-control study, in which 25 neonates were evaluated as the case group and 50 neonates as the control group, of both genders. DNA was extracted from peripheral blood leukocytes, and the site encompassing the polymorphism was amplified by molecular techniques (polymerase chain reaction/polymorphism in restriction fragment length). RESULTS Necrotizing enterocolitis was diagnosed in 25 (33%) neonates and, of these, 3 (12%) died. Male gender was more prevalent in both groups (p=0.1613): cases (52%) and controls (62%). Moderate and extreme preterm newborns were predominant in both groups: cases (80%) and controls (88%) (p=0.3036). Low birth weight and extremely low birth weight newborns were the most prevalent in cases (78%), and very low birth weight and extremely low birth weight were the most prevalent in controls (81%) (p=0.1073). Clinical treatment was successful in 72%, and hospital discharge was achieved in 88% of newborns with NEC. The -94ins/delATTG polymorphism in NFKB1 gene was not identified in all the 150 alleles analyzed (100%). CONCLUSIONS The absence of the -94ins/delATTG polymorphism in NFKB1 gene in newborns with and without necrotizing enterocolitis does not rule out the possibility of alterations in this and/or in other genes in newborns with this condition, which reinforces the need for further research.
Collapse
Affiliation(s)
| | | | - Vânia Belintani PIATTO
- Faculty of Medicine of São José do Rio Preto, Anatomy Unit – São José do Rio Preto (SP), Brazil
| | - Antônio Soares SOUZA
- Faculty of Medicine of São José do Rio Preto, Image Unit – São José do Rio Preto (SP), Brazil
| |
Collapse
|
82
|
Duess JW, Sampah ME, Lopez CM, Tsuboi K, Scheese DJ, Sodhi CP, Hackam DJ. Necrotizing enterocolitis, gut microbes, and sepsis. Gut Microbes 2023; 15:2221470. [PMID: 37312412 PMCID: PMC10269420 DOI: 10.1080/19490976.2023.2221470] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease in premature infants and the leading cause of death and disability from gastrointestinal disease in this vulnerable population. Although the pathophysiology of NEC remains incompletely understood, current thinking indicates that the disease develops in response to dietary and bacterial factors in the setting of a vulnerable host. As NEC progresses, intestinal perforation can result in serious infection with the development of overwhelming sepsis. In seeking to understand the mechanisms by which bacterial signaling on the intestinal epithelium can lead to NEC, we have shown that the gram-negative bacterial receptor toll-like receptor 4 is a critical regulator of NEC development, a finding that has been confirmed by many other groups. This review article provides recent findings on the interaction of microbial signaling, the immature immune system, intestinal ischemia, and systemic inflammation in the pathogenesis of NEC and the development of sepsis. We will also review promising therapeutic approaches that show efficacy in pre-clinical studies.
Collapse
Affiliation(s)
- Johannes W. Duess
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Maame E. Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Carla M. Lopez
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Koichi Tsuboi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Daniel J. Scheese
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Chhinder P. Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
83
|
Metzler M, Burns W, Mitchell C, Napolitano S, Chaudhari BP. A case report of necrotizing enterocolitis in a moderately preterm neonate with LCHADD-A call to focus on the basics while utilizing advanced new therapies. Front Pediatr 2023; 11:1081802. [PMID: 36861082 PMCID: PMC9969157 DOI: 10.3389/fped.2023.1081802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/16/2023] [Indexed: 02/15/2023] Open
Abstract
Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is an autosomal recessive condition of impaired beta-oxidation. Traditionally, treatment included restriction of dietary long-chain fatty acids via a low-fat diet and supplementation of medium chain triglycerides. In 2020, triheptanoin received FDA approval as an alternative source of medium chain fatty acids for individuals with long-chain fatty acid oxidation disorders (LC-FAOD). We present a case of a moderately preterm neonate born at 33 2/7 weeks gestational age with LCHADD who received triheptanoin and developed necrotizing enterocolitis (NEC). Prematurity is known as a major risk factor for NEC, with risk increasing with decreasing gestational age. To our knowledge, NEC has not previously been reported in patients with LCHADD or with triheptanoin use. While metabolic formula is part of the standard of care for LC-FAOD in early life, preterm neonates may benefit from more aggressive attempts to use skimmed human milk to minimize exposure to formula during the risk period for NEC during feed advancement. This risk period may be longer in neonates with LC-FAOD compared to otherwise healthy premature neonates.
Collapse
Affiliation(s)
- Marina Metzler
- Pediatric Residency, Nationwide Children's Hospital, Columbus, OH, United States
| | - William Burns
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Carly Mitchell
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Stephanie Napolitano
- Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, United States
| | - Bimal P Chaudhari
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, United States.,Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
84
|
Compositional Differences of Meconium Microbiomes of Preterm and Term Infants, and Infants That Developed Necrotizing Enterocolitis or Feeding Intolerance. Pathogens 2022; 12:pathogens12010055. [PMID: 36678403 PMCID: PMC9863680 DOI: 10.3390/pathogens12010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
The primary aim of this study was to investigate the compositional differences of the first passed meconium microbiome in preterm and term infants, and the secondary aim was to compare the meconium microbiomes of preterm and term infants that later developed necrotizing enterocolitis (NEC)/Feeding intolerance (FI) compared to those that did not develop NEC/FI. During the study period, a total of 108 preterm and term newborns' first passed meconium occurring within 72 hours of birth were collected and microbiome analyzed. Meconium microbiomes showed a disruption in the percentages of the core microbiome constituents in both the phylum and genus levels in infants born < 30 weeks of gestational age (GA) compared to those born ≥ 30 weeks of GA. In the phylum level, Bacteroidetes and Firmicutes, and in the genus level, Prevotella and Bacteroides, were predominant, with Prevotella accounting for 20−30% of the relative abundance. As GA increased, a significant increase in the relative abundance of Bacteroidetes (P for trend < 0.001) and decrease in Proteobacteria (P for trend = 0.049) was observed in the phylum level. In the genus level, as GA increased, Prevotella (P for trend < 0.001) and Bacteroides (P for trend = 0.002) increased significantly, whereas Enterococcus (P for trend = 0.020) decreased. Compared to the control group, the meconium of infants that later developed NEC/FI had significantly lower alpha diversities but similar beta-diversities. Furthermore, the NEC/FI group showed a significantly lower abundance of Bacteroidetes (P < 0.001), and higher abundance of Firmicutes (P = 0.034). To conclude, differences were observed in the composition of the first passed meconium in preterm and term infants that later develop NEC/FI compared to those that did not.
Collapse
|
85
|
CCL3 aggravates intestinal damage in NEC by promoting macrophage chemotaxis and M1 macrophage polarization. Pediatr Res 2022:10.1038/s41390-022-02409-w. [PMID: 36550354 DOI: 10.1038/s41390-022-02409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND NEC is a life-threatening gastrointestinal disease in neonates, the pathogenesis of which remains poorly understood. METHODS CCL3 levels in intestinal tissue of mice were measured and analyzed. HE staining was used to assess pathological changes in intestinal tissue. FCM was used to detect the proportion and phenotype of macrophages. RNA-seq and RT-PCR were used to evaluate the effect of CCL3 on macrophages. RESULTS CCL3 was highly expressed in the intestinal tissues of mice with NEC and induced macrophage infiltration. Transcriptome data showed that CCL3 strongly induced a transition in the phenotype of macrophages into a proinflammatory one. Mechanistically, in vivo experiments confirmed that CCL3 induced M1 macrophage polarization in NEC intestinal tissue, thereby aggravating inflammatory injury of intestinal tissue, which was alleviated by anti-CCL3 treatment. In addition, in vitro experiments showed that CCL3 significantly enhances the expression of M1-related genes in both PMφ and BMDM while inhibiting the expression of M2-related genes, which was also alleviated by anti-CCl3 treatment. CONCLUSIONS Our data elucidated the involvement of CCL3 in the pathogenesis of NEC, in which upregulated CCL3 expression exacerbated inflammatory intestinal damage by regulating macrophage chemotaxis and M1 phenotype polarization, suggesting that blocking CCL3 may be a potential strategy for effective intervention in NEC. IMPACT Our study represents an important conceptual advancement that CCL3 may be one of the key culprits of intestinal tissue damage in patients with NEC. CCL3 aggravates inflammatory intestinal injury and intestinal mucosal barrier imbalance by regulating the chemotaxis, polarization, and function of macrophages. Blocking CCL3 significantly reduced NEC-mediated intestinal injury, suggesting a new potential therapeutic strategy.
Collapse
|
86
|
Brown BE, Huisman E, Miller MR, Ulrich C, Reid G, da Silva O. Enteral supplementation with probiotics in preterm infants: A retrospective cohort study and 6-year follow-up. Front Nutr 2022; 9:1063121. [PMID: 36643974 PMCID: PMC9835551 DOI: 10.3389/fnut.2022.1063121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
The objective of this retrospective cohort study was to assess the impact of an enteral probiotics supplementation protocol on the incidence of necrotizing enterocolitis (NEC) in infants born <33 weeks gestational age (GA) or birth weight (BW) <1,500 g. In addition, a 6-year follow-up is presented after instigation of probiotic use. In October 2014, our NICU introduced an enteral probiotics supplementation protocol for infants born <33 weeks GA or BW <1,500 g. Infants received 0.5 g of Bifidobacterium breve HA-129, Lacticaseibacillus rhamnosus HA-111, Bifidobacterium bifidum HA-132, Bifidobacterium longum subsp. infantis HA-116, and Bifidobacterium longum subsp. longum HA-135 (FloraBABYⓇ) daily until discharge or transfer from hospital. The incidence of NEC was compared among infants for 2 years pre- and post implementation of the protocol then 6-years following continuous implementation of the probiotic use. In total, 370 infants not treated with probiotics between 2012 and 2014 were included with an incidence of NEC at 4.9%. In comparison, the 367 infants who received had a 67% reduction (4.9-1.6%, p = 0.01) in our Neonatal Intensive Care Unit (NICU). The results remained significant (aOR = 0.26; 95% CI: 0.09, 0.72; p < 0.01) after adjusting for GA, small for gestational age, and antenatal corticosteroid use. Data from the Canadian Neonatal Network not only showed a consistently high rate of NEC in October 2014, but also identified exceedingly high rates (8.7-15.6%) in some hospitals up to 2021, while our rates have been consistently low with using the probiotic as standard therapy for low BW premature babies, with no serious side effects reported. In conclusion, the introduction of a five-strain probiotic natural health product has coincided with a reduced incidence and complications of NEC in our NICU setting.
Collapse
Affiliation(s)
- Beth Ellen Brown
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Western Ontario, London, ON, Canada
| | - Esther Huisman
- Department of Pediatrics, Division of Neonatology, McMaster University, Hamilton, ON, Canada
| | - Michael R. Miller
- Department of Pediatrics, The Children's Health Research Institute, University of Western Ontario, London, ON, Canada
| | - Cindy Ulrich
- Neonatal Intensive Care Unit, Children's Hospital, London Health Sciences Centre, London, ON, Canada
| | - Gregor Reid
- Departments of Microbiology and Immunology, and Surgery, University of Western Ontario, London, ON, Canada,Canadian Centre for Microbiome and Probiotics, Lawson Health Research Institute, London, ON, Canada,*Correspondence: Gregor Reid
| | - Orlando da Silva
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Western Ontario, London, ON, Canada
| |
Collapse
|
87
|
Lactate acid level and prognosis of neonatal necrotizing enterocolitis: a retrospective cohort study based on pediatric-specific critical care database. J Pediatr (Rio J) 2022; 99:278-283. [PMID: 36535423 DOI: 10.1016/j.jped.2022.11.005] [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: 09/14/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To investigate the relationship between lactate acid level and hospitalization mortality in neonatal necrotizing enterocolitis (NEC). METHOD Paediatric-specific critical care database collected clinical data from the intensive care unit of Children's Hospital Affiliated to Zhejiang University Medical College from 2010 to 2018. Clinical and laboratory examination information of NEC patients was collected and divided into the death group and discharge group to find out the risk factors affecting the prognosis through univariate and multivariate analysis. RESULTS Among 104 NEC neonates, the admission age was 7.5 days and the weight was 2.03 kg. Comparing the death group with the discharge group, there were significant differences in therapeutic regimen, pH, serum albumin, total protein, creatinine and lactate acid. Multivariate and threshold effect analysis showed that lactate acid had a linear correlation with hospital mortality, and newborns who died in the hospital had much higher lactate levels than those who were discharged. The mortality of NEC newborns increased by 40-45% for every 1 mmol/L increase in lactate acid level. CONCLUSIONS There was a correlation between lactate acid level and hospital mortality in newborns with NEC, and lactate acid level was an important index to evaluate the prognosis of NEC.
Collapse
|
88
|
Sabbatini S, Ganji N, Chusilp S, Balsamo F, Li B, Pierro A. Intestinal atresia and necrotizing enterocolitis: Embryology and anatomy. Semin Pediatr Surg 2022; 31:151234. [PMID: 36417784 DOI: 10.1016/j.sempedsurg.2022.151234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The primitive gut originates at week 3 of gestation from the endoderm, with posterior incorporation of the remaining embryo layers. Wnt, Notch and TLR4 pathways have been shown to play central roles in the correct development of the intestine. The classical hypothesis for intestinal atresia development consists of failure in bowel recanalization or a vascular accident with secondary bowel reabsorption. These have been challenged due to the high frequency of associated malformations, and furthermore, with the discovery of molecular pathways and genes involved in bowel formation and correlated defects producing atresia. Necrotizing enterocolitis (NEC) has a multifactorial pathogenesis with prematurity being the most important risk factor; therefore, bowel immaturity plays a central role in NEC. Some of the same molecular pathways involved in gut maturation have been found to correlate with the predisposition of the immature bowel to develop the pathological findings seen in NEC.
Collapse
Affiliation(s)
- S Sabbatini
- Translational Medicine Program, The Hospital for Sick Children, Toronto
| | - N Ganji
- Translational Medicine Program, The Hospital for Sick Children, Toronto
| | - S Chusilp
- Translational Medicine Program, The Hospital for Sick Children, Toronto
| | - F Balsamo
- Translational Medicine Program, The Hospital for Sick Children, Toronto
| | - B Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto
| | - A Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto.
| |
Collapse
|
89
|
Sildenafil attenuates intestinal injury in necrotizing enterocolitis independently of endothelial nitric oxide synthase. J Pediatr Surg 2022; 57:967-973. [PMID: 35794042 DOI: 10.1016/j.jpedsurg.2022.06.001] [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: 03/07/2022] [Revised: 05/24/2022] [Accepted: 06/05/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating disease that impacts the intestine of premature infants. Sildenafil has shown benefit in colitis and ischemia/reperfusion models but has not been adequately studied in NEC. Sildenafil's best studied mechanism involves augmenting nitric oxide induced vasodilation. We hypothesized that sildenafil would improve outcomes during experimental NEC in an eNOS dependent manner. MATERIALS NEC was induced in five-day old mouse pups with gavage formula feeds plus intermittent hypoxia and hypothermia. Using wild type (WT) mice, the route of sildenafil administration was studied in the following groups: (1) breastfed controls, (2) NEC + oral (PO) sildenafil, (3) NEC + PO vehicle, (4) NEC + intraperitoneal (IP) sildenafil, (5) NEC + IP vehicle. The eNOS KO groups studied included: (1) breastfed controls, (2) NEC + PO sildenafil, (3) NEC + PO vehicle. Data were tested for normality and compared using t-tests or Mann-Whitney with a p-value <0.05 considered significant. RESULTS In WT mice, oral and IP sildenafil resulted in improved clinical outcomes compared to their respective vehicle group. Only orally administered sildenafil significantly improved perfusion to the intestine and protected it from macroscopic and histologic injury. When repeated in eNOS KO mice, oral sildenafil improved clinical scores and attenuated intestinal injury scores, despite no effect on intestinal perfusion. CONCLUSIONS Sildenafil, when administered orally, improves clinical outcomes and protects the intestine in a murine model of experimental necrotizing enterocolitis. While sildenafil requires eNOS to impact mesenteric perfusion, it does not appear to be dependent on eNOS to attenuate intestinal injury.
Collapse
|
90
|
Akduman H, Tayman C, Korkmaz V, Akduman F, Fettah ND, Gürsoy BK, Turkmenoglu TT, Çağlayan M. Astaxanthin Reduces the Severity of Intestinal Damage in a Neonatal Rat Model of Necrotizing Enterocolitis. Am J Perinatol 2022; 39:1820-1827. [PMID: 33853144 DOI: 10.1055/s-0041-1727156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE This study aimed to ascertain the effects of astaxanthin (ASX) in an experimental necrotizing enterocolitis (NEC) model using rat pups. STUDY DESIGN Forty-two pups born from five Wistar albino rats were randomly divided into three groups as the control group, NEC + placebo (saline), and NEC + ASX. Pups in the NEC + ASX group were given 100 mg/kg/day oral ASX from day 1 to day 4 of the study. Saline of 2 mL/kg was given to the NEC + placebo group. Histopathological, immunohistochemical (caspase-3), and biochemical evaluations including the total antioxidant status (TAS), total oxidant status (TOS), superoxide dismutase (SOD), glutathione (GSH), lipid hydroperoxide (LPO), 8-hydroxydeoxyguanosine (8-OHdG), advanced oxidation protein products (AOPP), myeloperoxidase (MPO), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and nuclear factor erythroid 2-related factor 2 (Nfr-2) activities were all performed. RESULTS A better survival rate and weight gain were demonstrated in the NEC + ASX group (p < 0.05). In the histopathological evaluation, the severity of intestinal damage was significantly reduced in the NEC + ASX group, as well as decreased apoptosis (enzyme-linked immunosorbent assay [ELISA] for caspase-3; p = 0.001). The biochemical analyses of intestinal tissue TOS, oxidative stress index (OSI; TOS/TAS), IL-1β, LPO, 8-OHdG, AOPP, caspase-3 (p < 0.001 for all), and TNF-α and MPO (p = 0.001 for both parameters) levels were lower in the NEC + ASX group than in the NEC + placebo group. Nrf-2, TAS, GSH, and SOD levels were higher in the NEC + ASX group than in the NEC + placebo group (p = 0.001, 0.001, <0.001, and 0.01, respectively). CONCLUSION ASX treatment has been shown to effectively reduce the severity of intestinal damage in NEC due to its antioxidant, anti-inflammatory, and antiapoptotic properties. KEY POINTS · NEC causes extremely high morbidity and mortality, as well as many complications.. · We investigated the effectiveness of ASX in the experimental NEC model created in rat pups.. · First study examining the effect of ASX on the experimental NEC rat model..
Collapse
Affiliation(s)
- Hasan Akduman
- Division of Neonatology, Department of Pediatrics, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Cuneyt Tayman
- Department of Neonatology, Ankara City Hospital, Cankaya, Ankara, Turkey
| | - Veli Korkmaz
- Department of Pediatrics, Lokman Hekim University, Ankara, Turkey
| | - Filiz Akduman
- Department of Pediatrics, Beypazarı State Hospital, Ankara, Turkey
| | - Nurdan D Fettah
- Department of Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Başak K Gürsoy
- Department of Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Tugba T Turkmenoglu
- Department of Pathology, Ankara Diskapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| | - Murat Çağlayan
- Department of Medicinal Biochemistry, University of Health Sciences Gülhane Health Sciences Institute, Ankara, Turkey
| |
Collapse
|
91
|
Predictors of Social-Emotional Development and Adaptive Functioning in School-Age Children with Intestinal Failure. J Clin Psychol Med Settings 2022:10.1007/s10880-022-09919-z. [PMID: 36371790 DOI: 10.1007/s10880-022-09919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Despite a focus on neurocognition in pediatric intestinal failure (IF) to date, we examined social-emotional and adaptive functioning. METHODS Children (N = 63) in our IF rehabilitation program underwent neuropsychological assessments including caregiver- and teacher-reported questionnaires. Results were compared to norms using z-tests. Caregiver and teacher reports were compared using t tests. Medical and demographic factors were examined in an exploratory manner using correlation and targeted regression analyses, adjusting for gestational age and full-scale IQ. RESULTS Caregiver and teacher reports indicated poorer executive, internalizing, behavioral, and adaptive functioning compared to norms. Teachers reported more executive dysfunctions than caregivers. Necrotizing enterocolitis diagnosis predicted internalizing emotional problems. Immigrant status predicted poorer social and practical adaptive functioning. Living with biological parents predicted fewer externalizing emotional and behavioral problems. CONCLUSIONS The group displayed social-emotional and adaptive functioning concerns. Identifying medical and demographic risks can allow for screening and intervention.
Collapse
|
92
|
Cerdó T, García-Santos JA, Rodríguez-Pöhnlein A, García-Ricobaraza M, Nieto-Ruíz A, G. Bermúdez M, Campoy C. Impact of Total Parenteral Nutrition on Gut Microbiota in Pediatric Population Suffering Intestinal Disorders. Nutrients 2022; 14:4691. [PMID: 36364953 PMCID: PMC9658482 DOI: 10.3390/nu14214691] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Parenteral nutrition (PN) is a life-saving therapy providing nutritional support in patients with digestive tract complications, particularly in preterm neonates due to their gut immaturity during the first postnatal weeks. Despite this, PN can also result in several gastrointestinal complications that are the cause or consequence of gut mucosal atrophy and gut microbiota dysbiosis, which may further aggravate gastrointestinal disorders. Consequently, the use of PN presents many unique challenges, notably in terms of the potential role of the gut microbiota on the functional and clinical outcomes associated with the long-term use of PN. In this review, we synthesize the current evidence on the effects of PN on gut microbiome in infants and children suffering from diverse gastrointestinal diseases, including necrotizing enterocolitis (NEC), short bowel syndrome (SBS) and subsequent intestinal failure, liver disease and inflammatory bowel disease (IBD). Moreover, we discuss the potential use of pre-, pro- and/or synbiotics as promising therapeutic strategies to reduce the risk of severe gastrointestinal disorders and mortality. The findings discussed here highlight the need for more well-designed studies, and harmonize the methods and its interpretation, which are critical to better understand the role of the gut microbiota in PN-related diseases and the development of efficient and personalized approaches based on pro- and/or prebiotics.
Collapse
Affiliation(s)
- Tomás Cerdó
- Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - José Antonio García-Santos
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Anna Rodríguez-Pöhnlein
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - María García-Ricobaraza
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Ana Nieto-Ruíz
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Mercedes G. Bermúdez
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada’s Node, Carlos III Health Institute, Avda. Monforte de Lemos 5, 28028 Madrid, Spain
| |
Collapse
|
93
|
Ahmad I, Premkumar MH, Hair AB, Sullivan KM, Zaniletti I, Sharma J, Nayak SP, Reber KM, Padula M, Brozanski B, DiGeronimo R, Yanowitz TD. Variability in antibiotic duration for necrotizing enterocolitis and outcomes in a large multicenter cohort. J Perinatol 2022; 42:1458-1464. [PMID: 35760891 DOI: 10.1038/s41372-022-01433-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 05/01/2022] [Accepted: 06/09/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVES To evaluate variability in antibiotic duration for necrotizing enterocolitis (NEC) and associated clinical outcomes. STUDY DESIGN Five-hundred ninety-one infants with NEC (315 medical; 276 surgical) were included from 22 centers participating in Children's Hospitals Neonatal Consortium (CHNC). Multivariable analyses were used to determine predictors of variability in time to full feeds (TFF) and length of stay (LOS). RESULTS Median (IQR) antibiotic duration was 12 (9, 17) days for medical and 17 (14, 21) days for surgical NEC. Wide variability in antibiotic use existed both within and among centers. Duration of antibiotic therapy was associated with longer TFF in both medical (OR 1.04, 95% CI [1.01, 1.05], p < 0.001) and surgical NEC (OR 1.02 [1, 1.03] p = 0.046); and with longer LOS in medical (OR 1.03 [1.02, 1.04], p < 0.001) and surgical NEC (OR 1.01 [1.01, 1.02], p = 0.002). CONCLUSION Antibiotic duration for both medical and surgical NEC remains variable within and among high level NICUs.
Collapse
Affiliation(s)
| | | | - Amy B Hair
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Kevin M Sullivan
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jotishna Sharma
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | | | - Kristina M Reber
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Michael Padula
- University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
94
|
Perego M, Fu S, Cao Y, Kossenkov A, Yao M, Bonner E, Alicea-Torres K, Liu W, Jiang Z, Chen Z, Fuchs SY, Zhou J, Gabrilovich DI. Mechanisms regulating transitory suppressive activity of neutrophils in newborns: PMNs-MDSCs in newborns. J Leukoc Biol 2022; 112:955-968. [PMID: 35726818 PMCID: PMC9794389 DOI: 10.1002/jlb.4hi0921-514rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/30/2022] Open
Abstract
Transitory appearance of immune suppressive polymorphonuclear neutrophils (PMNs) defined as myeloid-derived suppressor cells (PMNs-MDSCs) in newborns is important for their protection from inflammation associated with newly established gut microbiota. Here, we report that inhibition of the type I IFN (IFN1) pathway played a major role in regulation of PMNs-MDSCs-suppressive activity during first weeks of life. Expression of the IFN1 receptor IFNAR1 was markedly lower in PMNs-MDSCs. However, in newborn mice, down-regulation of IFNAR1 was not sufficient to render PMNs immune suppressive. That also required the presence of a positive signal from lactoferrin via its receptor low-density lipoprotein receptor-related protein 2. The latter effect was mediated via NF-κB activation, which was tempered by IFN1 in a manner that involved suppressor of cytokine signaling 3. Thus, we discovered a mechanism of tight regulation of immune suppressive PMNs-MDSCs in newborns, which may be used in the development of therapies of neonatal pathologies.
Collapse
Affiliation(s)
| | - Shuyu Fu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Yingjiao Cao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Meng Yao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Erin Bonner
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Alicea-Torres
- The Wistar Institute, Philadelphia, Pennsylvania, USA
- Biology Department, University of Puerto Rico-Humacao, Humacao, Puerto Rico, USA
| | - Wangkai Liu
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhilong Jiang
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | | |
Collapse
|
95
|
Tian B, Zhang Y, Deng C, Guo C. Efficacy of Probiotic Consortium Transplantation on Experimental Necrotizing Enterocolitis. J Surg Res 2022; 279:598-610. [PMID: 35926310 DOI: 10.1016/j.jss.2022.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/22/2022] [Accepted: 05/22/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Fecal microbiota transplantation (FMT) is a promising therapy, but it has not been used to treat neonatal necrotizing enterocolitis (NEC) due to reports of adverse side effects. Probiotics are considered relatively safe with practicable administrative procedures; however, no systematic research has compared the results of FMT and probiotic consortium transplantation (PCT) on oxidative stress in the intestines of patients with NEC. We conducted this study to provide a basis for optimizing NEC therapy. METHODS Eight-day-old newborn C57BL/6 mice were randomly divided into the following four groups: the dam-fed group (control group); the NEC induction group (NEC group); the NEC induction and transplantation of Lactobacillus reuteri and Bifidobacterium infantis consortium group (NEC + PCT group); and the NEC induction and the FMT group (NEC + FMT). Intestinal injury, oxidative stress indexes, intestinal barrier function, and inflammatory cytokines were assessed in the terminal ileum. RESULTS FMT more effectively modulates oxidative stress in the intestine than does PCT; however, the difference between the effects of PCT and FMT was not significant. The protective effect was associated with enhanced antioxidant capacity, regulation of the main components of the mucus layer, reduced inflammatory reactions, and improved intestinal integrity. CONCLUSIONS Intestinal dysbiosis affects oxidative stress, inflammatory response, and mucosal integrity. Although FMT is more effective than PCT in regulating oxidative stress, PCT may be preferred in pediatrics because the proportion and dose of transplanted bacteria can be standardized and individualized according to individual conditions.
Collapse
Affiliation(s)
- Bing Tian
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China; Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, Chongqin, China
| | - Yunfei Zhang
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China; School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China; National Clinical Research Center for Child Health and Disorders (Chongqing), Children's Hospital, Chongqing Medical University, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - Chun Deng
- Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, Chongqin, China; School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital, Chongqing Medical University, Chongqing, China.
| | - Chunbao Guo
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Children's Hospital, Chongqing Medical University, Chongqing, China; Department of Pediatric Surgery, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
96
|
Huang D, Wang P, Chen J, Li Y, Zhu M, Tang Y, Zhou W. Selective targeting of MD2 attenuates intestinal inflammation and prevents neonatal necrotizing enterocolitis by suppressing TLR4 signaling. Front Immunol 2022; 13:995791. [PMID: 36389716 PMCID: PMC9663461 DOI: 10.3389/fimmu.2022.995791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/14/2022] [Indexed: 10/17/2023] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is an inflammatory disease that occurs in premature infants and has a high mortality rate; however, the mechanisms behind this disease remain unclear. The TLR4 signaling pathway in intestinal epithelial cells, mediated by TLR4, is important for the activation of the inflammatory storm in NEC infants. Myeloid differentiation protein 2 (MD2) is a key auxiliary component of the TLR4 signaling pathway. In this study, MD2 was found to be significantly increased in intestinal tissues of NEC patients at the acute stage. We further confirmed that MD2 was upregulated in NEC rats. MD2 inhibitor (MI) pretreatment reduced the occurrence and severity of NEC in neonatal rats, inhibited the activation of NF-κB and the release of inflammatory molecules (TNF-α and IL-6), and reduced the severity of intestinal injury. MI pretreatment significantly reduced enterocyte apoptosis while also maintaining tight junction proteins, including occludin and claudin-1, and protecting intestinal mucosal permeability in NEC rats. In addition, an NEC in vitro model was established by stimulating IEC-6 enterocytes with LPS. MD2 overexpression in IEC-6 enterocytes significantly activated NF-κB. Further, both MD2 silencing and MI pretreatment inhibited the inflammatory response. Overexpression of MD2 increased damage to the IEC-6 monolayer cell barrier, while both MD2 silencing and MI pretreatment played a protective role. In conclusion, MD2 triggers an inflammatory response through the TLR4 signaling pathway, leading to intestinal mucosal injury in NEC. In addition, MI alleviates inflammation and reduces intestinal mucosal injury caused by the inflammatory response by blocking the TLR4-MD2/NF-κB signaling axis. These results suggest that inhibiting MD2 may be an important way to prevent NEC.
Collapse
Affiliation(s)
- Dabin Huang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ping Wang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Juncao Chen
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanbin Li
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaping Tang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhou
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
97
|
Dang D, Zhang C, Meng Z, Lv X, Li Z, Wei J, Wu H. Integrative analysis links ferroptosis to necrotizing enterocolitis and reveals the role of ACSL4 in immune disorders. iScience 2022; 25:105406. [DOI: 10.1016/j.isci.2022.105406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/24/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
|
98
|
Han Y, Liu X, Kang L, Chen D, Li Y, Zhang H, Sun M, Gao H, Gai Z, Li X. A potential pathogenic hypoxia-related gene HK2 in necrotizing enterocolitis (NEC) of newborns. BMC Pediatr 2022; 22:617. [PMID: 36289463 PMCID: PMC9597967 DOI: 10.1186/s12887-022-03664-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a disastrous gastrointestinal disease of newborns, and the mortality rate of infants with NEC is approximately 20%-30%. The exploration of pathogenic targets of NEC will be conducive to timely diagnosis of NEC. Methods The whole transcriptome RNA sequencing was performed on NEC samples to reveal the expression of lncRNAs, circRNAs, miRNAs and mRNAs. Using differential expression analysis, cross analysis, target prediction, enrichment analysis, the pathogenic ceRNA network and target was found. Results Preliminarily, 281 DEmRNAs, 21 DEmiRNAs, 253 DElncRNAs and 207 DEcircRNAs were identified in NEC samples compared with controls. After target prediction and cross analyses, a key ceRNA regulatory network was built including 2 lncRNAs, 4 circRNAs, 2 miRNAs and 20 mRNAs. These 20 mRNAs were significantly enriched in many carbohydrate metabolism related pathways. After cross analysis of hypoxia-, carbohydrate metabolism-related genes, and 20 core genes, one gene HK2 was finally obtained. Dendritic cells activated were significantly differentially infiltrated and negatively correlated with HK2 expression in NEC samples. Conclusions The promising pathogenic hypoxia-related gene HK2 has been firstly identified in NEC, which might also involve in the carbohydrate metabolism in NEC. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03664-w.
Collapse
Affiliation(s)
- Yujie Han
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Xianghong Liu
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Lili Kang
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Dong Chen
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Yongqing Li
- Department of Neonatal, LaoLing Maternity and Child Health Care Hospital, 118 Anju Road, Laoling County, Dezhou, Shandong Province, 253600 People’s Republic of China
| | - Huiping Zhang
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Mingying Sun
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Hui Gao
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Zhongtao Gai
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Xiaoying Li
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| |
Collapse
|
99
|
Gagné D, Shajari E, Thibault MP, Noël JF, Boisvert FM, Babakissa C, Levy E, Gagnon H, Brunet MA, Grynspan D, Ferretti E, Bertelle V, Beaulieu JF. Proteomics Profiling of Stool Samples from Preterm Neonates with SWATH/DIA Mass Spectrometry for Predicting Necrotizing Enterocolitis. Int J Mol Sci 2022; 23:ijms231911601. [PMID: 36232903 PMCID: PMC9569884 DOI: 10.3390/ijms231911601] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening condition for premature infants in neonatal intensive care units. Finding indicators that can predict NEC development before symptoms appear would provide more time to apply targeted interventions. In this study, stools from 132 very-low-birth-weight (VLBW) infants were collected daily in the context of a multi-center prospective study aimed at investigating the potential of fecal biomarkers for NEC prediction using proteomics technology. Eight of the VLBW infants received a stage-3 NEC diagnosis. Stools collected from the NEC infants up to 10 days before their diagnosis were available for seven of them. Their samples were matched with those from seven pairs of non-NEC controls. The samples were processed for liquid chromatography-tandem mass spectrometry analysis using SWATH/DIA acquisition and cross-compatible proteomic software to perform label-free quantification. ROC curve and principal component analyses were used to explore discriminating information and to evaluate candidate protein markers. A series of 36 proteins showed the most efficient capacity with a signature that predicted all seven NEC infants at least a week in advance. Overall, our study demonstrates that multiplexed proteomic signature detection constitutes a promising approach for the early detection of NEC development in premature infants.
Collapse
Affiliation(s)
- David Gagné
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Elmira Shajari
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Marie-Pier Thibault
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Noël
- PhenoSwitch Bioscience Inc., 975 Rue Léon-Trépanier, Sherbrooke, QC J1G 5J6, Canada
| | - François-Michel Boisvert
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Corentin Babakissa
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Emile Levy
- Research Center, Centre Hospitalier Universitaire Ste-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Hugo Gagnon
- PhenoSwitch Bioscience Inc., 975 Rue Léon-Trépanier, Sherbrooke, QC J1G 5J6, Canada
| | - Marie A. Brunet
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - David Grynspan
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Colombia, Vancouver, BC V6T 2B5, Canada
| | - Emanuela Ferretti
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Valérie Bertelle
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
100
|
The administration of a pre-digested fat-enriched formula prevents necrotising enterocolitis-induced lung injury in mice. Br J Nutr 2022; 128:1050-1063. [PMID: 34632971 PMCID: PMC8995403 DOI: 10.1017/s0007114521004104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Necrotising enterocolitis (NEC) is a devastating gastrointestinal disease of prematurity that typically develops after the administration of infant formula, suggesting a link between nutritional components and disease development. One of the most significant complications that develops in patients with NEC is severe lung injury. We have previously shown that the administration of a nutritional formula that is enriched in pre-digested Triacylglyceride that do not require lipase action can significantly reduce the severity of NEC in a mouse model. We now hypothesise that this 'pre-digested fat (PDF) system' may reduce NEC-associated lung injury. In support of this hypothesis, we now show that rearing newborn mice on a nutritional formula based on the 'PDF system' promotes lung development, as evidenced by increased tight junctions and surfactant protein expression. Mice that were administered this 'PDF system' were significantly less vulnerable to the development of NEC-induced lung inflammation, and the administration of the 'PDF system' conferred lung protection. In seeking to define the mechanisms involved, the administration of the 'PDF system' significantly enhanced lung maturation and reduced the production of reactive oxygen species (ROS). These findings suggest that the PDF system protects the development of NEC-induced lung injury through effects on lung maturation and reduced ROS in the lung and also increases lung maturation in non-NEC mice.
Collapse
|