1
|
Shah S. The "pro" and "con" of probiotics, regulation and preterm infant health. Pediatr Res 2024:10.1038/s41390-024-03766-4. [PMID: 39633020 DOI: 10.1038/s41390-024-03766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Shetal Shah
- Division of Neonatology, Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
2
|
Zheng Y, Fowler J, Rector C, Tumin D, Herco M. Impact of changing donor human milk feeding guideline for extremely preterm infants on the use of infant formula and cost of donor human milk purchase. J Perinatol 2024:10.1038/s41372-024-02182-0. [PMID: 39567648 DOI: 10.1038/s41372-024-02182-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/10/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Affiliation(s)
- Yingying Zheng
- Department of Neonatology, East Carolina University Health Medical Center, Greenville, NC, USA
| | - Jennifer Fowler
- Department of Clinical Nutrition Services, East Carolina University Health Medical Center, Greenville, NC, USA
| | - Chance Rector
- Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Dmitry Tumin
- Department of Pediatrics, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Maja Herco
- Department of Neonatology, East Carolina University Health Medical Center, Greenville, NC, USA.
| |
Collapse
|
3
|
Tefera BA, Ahmed AM, Yehualashet SS. Time to recovery from necrotizing enterocolitis and its predictors among neonates admitted to Neonatal Intensive Care Unit in Bahir Dar, Ethiopia: A retrospective follow up study, 2022. PLoS One 2024; 19:e0311890. [PMID: 39436945 PMCID: PMC11495620 DOI: 10.1371/journal.pone.0311890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Necrotizing enterocolitis is one of the most common, life-threatening, gastrointestinal disorders in neonates. The recovery time for neonates with NEC varies depending on disease severity, prompt diagnosis, and effective treatment. Therefore, this study was intended to assess the time to recover from necrotizing enterocolitis and its' predictors among neonates admitted to Neonatal Intensive Care Unit in Bahir Dar City, Ethiopia. METHODS An institution-based retrospective follow-up study design was employed. A sample of 361 medical records of neonates with necrotizing enterocolitis was selected using systematic random sampling. Diagnosis of NEC in this study required clinical, laboratory and radiographic findings. The survival function was described using Kaplan Meier survival curve and log-rank test. Bivariate and multivariate Cox-proportional hazard (Cox-PH) regression models were used for analysis. RESULTS The median recovery time from necrotizing enterocolitis for neonates in the neonatal intensive care unit was 12 days. The multivariable Cox-PH model showed that neonates classified as Stage III NEC (AHR: 0.42, 95% CI = 0.23-0.77) and those exposed to perinatal asphyxia (AHR: 0.51, 95% CI: 0.35-0.74) had a negative impact on NEC recovery time. However, neonates with a birth weight of 1500-2499gm (AHR: 1.65, 95% CI: 1.05-2.58) and a platelet count greater than 150,000 (AHR: 1.75, 95% CI: 1.24-2.48) had a positive effect on NEC recovery time. CONCLUSION The recovery time for neonates in the neonatal intensive care unit with necrotizing enterocolitis was longer. Comorbidities and advanced stage of NEC were associated with prolonged recovery time from NEC. However, neonates with better platelet count and birth weight greater than 1500mg had shorter recovery time from NEC.
Collapse
Affiliation(s)
- Birtukan Ayana Tefera
- Neonatal Intensive Care Unit, Felege Hiwot Comprehensive Specialized Hospital, Bahir Dar, Amhara Regional State, Ethiopia
| | - Abdurahman Mohammed Ahmed
- School of Public Health, Asrat Woldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Amhara Regional State, Ethiopia
| | - Sisay Shewasinad Yehualashet
- Department of Paediatrics and Child Health Nursing, School of Nursing and Midwifery, Asrat Woldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Amhara Regional State, Ethiopia
| |
Collapse
|
4
|
Zhang L, Jin W, Hu M, Su Y, Zhang Y, Yuan F, Fang Y, Li Z, Li Y, Bu C, Zhou W. Silencing miR-155-5p expression improves intestinal damage through inhibiting inflammation and ferroptosis in necrotizing enterocolitis. Heliyon 2024; 10:e37087. [PMID: 39286078 PMCID: PMC11402723 DOI: 10.1016/j.heliyon.2024.e37087] [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: 04/09/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a condition characterized by acquired damage to the mucosal lining, predominantly affecting premature infants. Bioinformatics assessments uncovered a notable rise in miR-155-5p expression in the intestinal tissues of infants suffering from NEC. Nevertheless, the development of NEC's underlying mechanisms and the role of miR-155-5p are still not well understood. This research aimed to explore the role of miR-155-5p in NEC and to elucidate its underlying mechanisms. Methods To replicate NEC in vitro, lipopolysaccharide (LPS) was employed, whereas an in vivo rat model of NEC was established using formula feeding and exposure to hypoxia. Subsequently, levels of inflammatory cytokines, cell survival, and apoptosis rates were assessed. Various biochemical indicators such as glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and malondialdehyde (MDA) were measured utilizing a purchased diagnostic kit. For the assessment of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) within FHC cells, analysis by flow cytometry was conducted. Additionally, the technique of Western blotting was utilized to analyze the levels of ferroptosis-associated proteins. Moreover, hematoxylin and eosin (H&E) staining was carried out to observe the histopathological alterations in the intestinal tissue samples from rats with necrotizing enterocolitis (NEC). Results Reducing miR-155-5p improved the survival of FHC cells exposed to LPS, decreased cell apoptosis, inflammation, and ferroptosis, and mitigated intestinal damage in NEC rats. Furthermore, SLC7A11 was found to be a direct target of miR-155-5p. The inhibition of miR-155-5p decreased LPS-induced inflammation and ferroptosis in both FHC cells and NEC rats by promoting SLC7A11 expression. This effect was evidenced by increased levels of ferroptosis-related proteins FTH1 and GPX4, decreased COX-2 and ACSL4 levels, lower lipid peroxidation marker MDA, reduced antioxidant markers GSH, SOD, and CAT, fewer IL-6 and TNF-α, and suppression of the IκBα/NF-κB p65 signaling pathway. Conclusions In conclusion, reducing miR-155-5p could improve intestinal damage in NEC by inhibiting inflammation and ferroptosis. These findings may provide theoretical insights for the development of new therapies for NEC.
Collapse
Affiliation(s)
- Le Zhang
- Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Weilai Jin
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Mengyuan Hu
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Yinglin Su
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Yiting Zhang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Fuqiang Yuan
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Yuanyuan Fang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Zhengying Li
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Yawen Li
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, Jiangsu, China
| | - Chaozhi Bu
- State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Wenhao Zhou
- Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Gerçel G, Anadolulu AI. Neonatal Gastrointestinal Perforations: A 4-year Experience in a Single Centre. Afr J Paediatr Surg 2024:01434821-990000000-00016. [PMID: 39254062 DOI: 10.4103/ajps.ajps_96_23] [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: 08/06/2023] [Accepted: 01/05/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Gastrointestinal perforation (GIP) during the neonatal period is still a significant problem despite improved neonatal care. The study aimed to report on incidence, management, morbidity and mortality. MATERIAL AND METHODS Records of neonates with GIPs between October 2018 and November 2022 were retrospectively analysed. RESULTS There were 47 patients, 22 (46.8%) males and 25 (53.2%) females. The incidence of neonatal GIP was 0.39% amongst all newborns treated in the neonatal intensive care unit. The mean gestational age was 30.4 ± 4.5 (23-38) weeks, and the mean birth weight was 1493.08 ± 753 (580-2940) g. Of 47 neonates, 5 (10.6%) were full term and 42 (89.4%) were preterm. The mean age of surgery was 12.25 ± 9.89 (0-41) days. A laparotomy was performed in 43 (91.4%) of 47 neonates, while seven of the patients underwent surgical intervention after decompression by percutaneous drainage. Four patients were managed with peritoneal drainage alone due to poor general condition. The pathologies unrelated to necrotising enterocolitis (NEC) were the most common cause of GIPs (55.3%) and included spontaneous intestinal perforation (n = 18), stomach perforation (n = 4), segmental volvulus (n = 2), acute mesenteric ischaemia (n = 1) and meconium peritonitis (n = 1). Overall survival was 55.4%. CONCLUSION GIPs are one of the most significant causes of mortality in newborns. The most common cause of perforations is non-NEC entities and can be seen in the entire intestinal system from the stomach to the colon. Surgical exploration is still the primary management model.
Collapse
Affiliation(s)
- Gonca Gerçel
- Department of Pediatric Surgery, Şanlıurfa Training and Research Hospital, Şanlıurfa, Turkey
- Department of Pediatric Surgery, İstanbul Sancaktepe Şehit Prof. Dr. İlhan Varank Training and Research Hospital, Istanbul, Turkey
| | - Ali Ihsan Anadolulu
- Department of Pediatric Surgery, Mehmet Akif İnan Training and Research Hospital, Şanlıurfa, Turkey
- Department of Pediatric Surgery, Faculty of Medicine, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
6
|
Dantes G, Murfee J, Doll A, Weaver K, Alemayehu H. Weight at Ostomy Takedown as a Factor to Consider for Operative Timing-Is It Relevant? J Laparoendosc Adv Surg Tech A 2024; 34:855-860. [PMID: 39162564 DOI: 10.1089/lap.2024.0188] [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: 08/21/2024] Open
Abstract
Purpose: Weight thresholds have historically determined timing of enterostomy closure (EC) in premature neonates. Recent evidence suggests that neonates less than 2 kg (L2K) can safely undergo EC. We evaluate our single-center experience with performing EC in preterm neonates at L2K versus greater than 2 kg (G2K) at time of EC. Methods: A retrospective review of neonates who underwent EC from January 2018 to 2020 was performed. Neonates who were greater than 90 days at initial operation were excluded. Demographics, clinical characteristics including gestational age (GA) and birth weight (BW), operative reports, and outcomes were reviewed. We compared 30-day complications between neonates who underwent EC at L2K and G2K. We also compared time to full feeds (FF) and postoperative length of stay (LOS). Results: Twenty-four neonates were included: 11 L2K and 13 G2K. The median GA and BW was 25.9 weeks (IQR 2.89) and 805 g (IQR 327), respectively. The most common intraoperative diagnosis during index operation was spontaneous perforation (70%), followed by necrotizing enterocolitis (8.69%). There were no significant differences in GA, BW, or diagnosis, between the L2K versus G2K cohort. We found no difference in complication rates, time to FF (12 days versus 10 days, P = .89), or postoperative LOS (31 days versus 36.5 days, P = .76) between patients who underwent EC at L2K versus G2K, respectively. Conclusion: Although weight gain may be an important indicator of perioperative nutrition status, this study shows that weight alone should not preclude otherwise appropriate patients from undergoing EC.
Collapse
Affiliation(s)
- Goeto Dantes
- Department of Surgery, Division of Pediatric Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jack Murfee
- Department of Surgery, Division of Pediatric Surgery, University of South Alabama, Mobile, Alabama, USA
| | - Alissa Doll
- Department of Surgery, Division of Pediatric Surgery, University of South Alabama, Mobile, Alabama, USA
| | - Katrina Weaver
- Department of Surgery, Division of Pediatric Surgery, University of South Alabama, Mobile, Alabama, USA
| | - Hanna Alemayehu
- Department of Surgery, Division of Pediatric Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Blomqvist YT, Olsson E. Experiences of breast milk donors in Sweden: balancing the motivation to do something good with overcoming the challenges it entails. Int Breastfeed J 2024; 19:60. [PMID: 39217315 PMCID: PMC11365258 DOI: 10.1186/s13006-024-00668-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/24/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Infants requiring neonatal care often face initial breastfeeding challenges, leading them to receive expressed breast milk from their mother or donor milk. While emphasizing the mother's own milk as the gold standard for infant nutrition, the utilization of donor milk stands as the preferred alternative over infant formula due to its numerous benefits. To facilitate the provision of donor milk to preterm and ill infants in neonatal units, the active participation of women willing to contribute their breast milk is crucial. This study aims to enhance the understanding of women's experiences in the donation process, thereby contributing to efforts aiming at alleviating the shortage of donated breast milk by improve the care and support for breast milk donors. METHODS This descriptive qualitative study took an inductive approach based on individual semi-structured interviews conducted during 2021 with 15 breast milk donors in Sweden. The data were analysed with thematic analysis. RESULTS Two themes were identified in the analysis: motivation to donate and challenges to overcome. Many of the women struggled to overcome the apparent challenges of not only starting the process of donating breast milk but also maintaining it. Despite the strain, they were motivated to donate their breast milk and seeking information by themselves to do something important for someone else. Only a few of the women talked about the financial benefits of donating breast milk; donating seemed to be mostly based on altruistic reasons. CONCLUSIONS Despite the challenges posed by COVID-19 restrictions, time consumption, and the hard work of sterilizing pump utensils, women continued to donate their milk driven by altruism. To enhance donor support and increase milk donation, several improvements are suggested: providing comprehensive information and resources, simplifying the donation process, offering flexible scheduling, and recognizing donors' contributions.
Collapse
Affiliation(s)
- Ylva Thernström Blomqvist
- Neonatal Intensive Care Unit, University Children's Hospital, Uppsala, Sweden.
- Department of Women's and Children's Health, Uppsala University, Uppsala, 751 85, Sweden.
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Faculty of Medicine and Health, School of Health Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
8
|
Nardini P, Zizi V, Molino M, Fazi C, Calvani M, Carrozzo F, Giuseppetti G, Calosi L, Guasti D, Biagini D, Di Francesco F, Filippi L, Pini A. Protective Effects of Beta-3 Adrenoceptor Agonism on Mucosal Integrity in Hyperoxia-Induced Ileal Alterations. Antioxidants (Basel) 2024; 13:863. [PMID: 39061931 PMCID: PMC11273805 DOI: 10.3390/antiox13070863] [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: 06/19/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Organogenesis occurs in the uterus under low oxygen levels (4%). Preterm birth exposes immature newborns to a hyperoxic environment, which can induce a massive production of reactive oxygen species and potentially affect organ development, leading to diseases such as necrotizing enterocolitis. The β3-adrenoreceptor (β3-AR) has an oxygen-dependent regulatory mechanism, and its activation exerts an antioxidant effect. To test the hypothesis that β3-AR could protect postnatal ileal development from the negative impact of high oxygen levels, Sprague-Dawley rat pups were raised under normoxia (21%) or hyperoxia (85%) for the first 2 weeks after birth and treated or not with BRL37344, a selective β3-AR agonist, at 1, 3, or 6 mg/kg. Hyperoxia alters ileal mucosal morphology, leading to increased cell lipid oxidation byproducts, reduced presence of β3-AR-positive resident cells, decreased junctional protein expression, disrupted brush border, mucin over-production, and impaired vascularization. Treatment with 3 mg/kg of BRL37344 prevented these alterations, although not completely, while the lower 1 mg/kg dose was ineffective, and the higher 6 mg/kg dose was toxic. Our findings indicate the potential of β3-AR agonism as a new therapeutic approach to counteract the hyperoxia-induced ileal alterations and, more generally, the disorders of prematurity related to supra-physiologic oxygen exposure.
Collapse
Affiliation(s)
- Patrizia Nardini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
- Imaging Platform, Department Experimental and Clinical Medicine & Joint Laboratory with Department Biology, University of Florence, 50139 Florence, Italy
| | - Virginia Zizi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
| | - Marta Molino
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
| | - Camilla Fazi
- Department of Pediatric, Meyer Children’s University Hospital, 50139 Florence, Italy;
| | - Maura Calvani
- Azienda Ospedaliera Universitaria Meyer, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 50139 Florence, Italy;
| | - Francesco Carrozzo
- Department of Health Science, University of Florence, 50139 Florence, Italy;
| | - Giorgia Giuseppetti
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
| | - Laura Calosi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
- Imaging Platform, Department Experimental and Clinical Medicine & Joint Laboratory with Department Biology, University of Florence, 50139 Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
- Imaging Platform, Department Experimental and Clinical Medicine & Joint Laboratory with Department Biology, University of Florence, 50139 Florence, Italy
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy; (D.B.); (F.D.F.)
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy; (D.B.); (F.D.F.)
| | - Luca Filippi
- Neonatology and Neonatal Intensive Care Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
| | - Alessandro Pini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (G.G.); (L.C.); (D.G.)
- Imaging Platform, Department Experimental and Clinical Medicine & Joint Laboratory with Department Biology, University of Florence, 50139 Florence, Italy
| |
Collapse
|
9
|
Guo H, Li Y, Wang L. Assessment of inflammatory biomarkers to identify surgical/death necrotizing enterocolitis in preterm infants without pneumoperitoneum. Pediatr Surg Int 2024; 40:191. [PMID: 39012349 DOI: 10.1007/s00383-024-05787-w] [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: 07/10/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a life-threatening disease that affects premature infants. However, the role of inflammatory biomarkers in identifying surgical/death NEC without pneumoperitoneum remains elusive. PURPOSE We aimed to verify the value of platelet-to-lymphocyte ratio (PLR) and the combination of white blood cell (WBC), absolute neutrophil count (ANC), absolute lymphocyte count (ALC), neutrophil lymphocyte ratio (NLR), PLR, C reactive protein (CRP) and procalcitonin (PCT) in predicting the severity of NEC, and to construct a model to differ surgically NEC from non-surgically NEC. METHODS A retrospective analysis was performed on 191 premature infants with NEC. Based on the inclusion and exclusion criteria, 90 infants with Stage II and IIIA NEC were enrolled in this study, including surgical/death NEC (n = 38) and medical NEC (n = 52). The values of inflammatory biomarkers were collected within 24 h of onset. RESULTS The univariate analysis revealed that the values of WBC (p = 0.040), ANC (p = 0.048), PLR (p = 0.009), CRP (p = 0.016) and PCT (p < 0.01) in surgical/death NEC cohort were significantly higher than medical NEC cohort. Binary multivariate logistic regression analysis indicates that ANC, PLR, CRP, and PCT are capable of distinguishing infants with surgical/death NEC, and the AUC of the regression equation was 0.79 (95% CI 0.64-0.89; sensitivity 0.63; specificity 0.88), suggesting the equation has a good discrimination. IMPLICATIONS FOR PRACTICE AND RESEARCH Elevated PLR is associated with severe inflammation in surgical/death NEC patients. The prediction modelling of combination of ANC, PLR, CRP and PCT can differentiate surgical/death NEC from infants with medical NEC, which may improve risk awareness and facilitate effective communication between nurses and clinicians. However, multicentre research is needed to verify these findings for better clinical management of NEC.
Collapse
MESH Headings
- Humans
- Enterocolitis, Necrotizing/surgery
- Enterocolitis, Necrotizing/blood
- Enterocolitis, Necrotizing/diagnosis
- Retrospective Studies
- Infant, Newborn
- Biomarkers/blood
- Male
- Female
- Infant, Premature
- C-Reactive Protein/analysis
- Procalcitonin/blood
- Pneumoperitoneum/blood
- Inflammation/blood
- Leukocyte Count
- Infant, Premature, Diseases/surgery
- Infant, Premature, Diseases/blood
- Infant, Premature, Diseases/diagnosis
Collapse
Affiliation(s)
- Haiyan Guo
- Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
- Department of Paediatrics, The First Affiliated Hospital of Anhui Medical University, Anhui Province, Hefei City, 230022, People's Republic of China
| | - Yuanzhi Li
- Department of Paediatrics, The First Affiliated Hospital of Anhui Medical University, Anhui Province, Hefei City, 230022, People's Republic of China
| | - Lili Wang
- Department of Paediatrics, The First Affiliated Hospital of Anhui Medical University, Anhui Province, Hefei City, 230022, People's Republic of China.
| |
Collapse
|
10
|
Rebai N, Lopriore E, Bekker V, Slaghekke F, Schoenaker MHD, Groene SG. Necrotizing enterocolitis in monochorionic twins: Insights from an identical twin model. Early Hum Dev 2024; 194:106052. [PMID: 38781714 DOI: 10.1016/j.earlhumdev.2024.106052] [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] [Received: 03/25/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of neonatal morbidity and mortality in preterm neonates, yet its pathophysiology remains unclear. The aim of this study is to evaluate risk factors for NEC using an identical twin model. In this case-control study, all monochorionic twin pairs born in our center in 2002-2020 were retrospectively reviewed for NEC. Potential risk factors for NEC were studied. For within-pair comparison, outcomes were compared between affected and unaffected twins. Within-pair analyses showed that the twin with NEC had a lower birth weight compared to its unaffected co-twin (1100 (913-1364) vs. 1339 (1093-1755) grams). Median gestational age at birth and birth weight were lower in twin pairs in the NEC-group compared to the no-NEC group, 29.1 weeks (27.8-30.8) versus 33.6 (30.7-36.0) and 1221 g (1010-1488) versus 1865 (1356-2355) respectively. Twin pregnancies in the NEC-group were more often complicated by twin-to-twin transfusion syndrome compared to the no-NEC-group (70 % (14/20) vs. 49 % (472/962)), particularly when treated with amnioreduction. This unique population of identical twins confirms that preterm neonates with a relatively lower birth weight are more prone to develop NEC compared to their co-twin, regardless of other genetic, maternal and obstetrical factors.
Collapse
Affiliation(s)
- Nour Rebai
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Enrico Lopriore
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Vincent Bekker
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Femke Slaghekke
- Department of Obstetrics, Division of Fetal Therapy, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Michiel H D Schoenaker
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Willem-Alexander Children's Hospital, Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Sophie G Groene
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
11
|
Rana M, Vega Gonzales-Portillo JD, Hahn C, Dutt M, Sanchez-Fernandez I, Jonas R, Douglass L, Torres AR. Current Evidence: Seizures in Extremely Low Gestational Age Newborns (ELGANs). J Child Neurol 2024; 39:285-291. [PMID: 38836290 DOI: 10.1177/08830738241259052] [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: 06/06/2024]
Abstract
Extremely low gestational age newborns (ELGANs) are born at or below 28 weeks of gestational age. Despite improved obstetric care, the incidence of preterm birth continues to rise in advanced countries. Preterm birth remains a major cause of infant mortality, and for infants who survive, neonatal seizures are a significant predictor of later neurologic morbidity. However, little is known about risk factors for neonatal seizures in ELGANs. Understanding the association between neonatal seizures and the development of other neurologic disorders is important given the increasing prevalence of ELGANs. Identifying risk factors that contribute to the development of neonatal seizures in ELGANs may offer insights into novel mechanisms of epileptogenesis in the developing brain and improvements in the prevention or treatment of seizures in preterm infants, including ELGANs. In this literature review, we outline the limitations of epidemiologic studies of neonatal seizures in ELGANs and discuss risk factors for neonatal seizures.
Collapse
Affiliation(s)
- Mandeep Rana
- Division of Pediatric Neurology and Sleep Medicine, Department of Pediatrics, Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Juan Diego Vega Gonzales-Portillo
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Cecil Hahn
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Monideep Dutt
- Division of Pediatric Neurology, Children's Healthcare of Atlanta: Pediatric Institute, Emory University, Atlanta, GA, USA
| | - Ivan Sanchez-Fernandez
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Rinat Jonas
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Laurie Douglass
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alcy R Torres
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
12
|
Jirillo E, Topi S, Charitos IA, Santacroce L, Gaxhja E, Colella M. Gut Microbiota and Immune System in Necrotizing Enterocolitis and Related Sepsis. GASTROINTESTINAL DISORDERS 2024; 6:431-445. [DOI: 10.3390/gidisord6020029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
A severe condition of sepsis can be a complication of necrotizing enterocolitis (NEC), which can occur in premature infants and becomes a medical challenge in the neonatal intensive care unit (NICU). It is a multifactorial intestinal disease (can affect both the small and large intestine) that can lead to ischemia of the intestinal tissues that evolves into acute organ necrosis. One of these factors is that different types of nutrition can influence the onset or the progression of the disease. Cow-milk-based infant formulas have been shown to cause it in premature infants more frequently than human milk. Recently, nutrition has been shown to be beneficial after surgery. Several issues still under study, such as the pathogenesis and the insufficient and often difficult therapeutic approach, as well as the lack of a common and effective prevention strategy, make this disease an enigma in daily clinical practice. Recent studies outlined the emerging role of the host immune system and resident gut microbiota, showing their close connection in NEC pathophysiology. In its initial stages, broad-spectrum antibiotics, bowel rest, and breastfeeding are currently used, as well as probiotics to help the development of the intestinal microbiota and its eubiosis. This paper aims to present the current knowledge and potential fields of research in NEC pathophysiology and therapeutic assessment.
Collapse
Affiliation(s)
- Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
| | - Skender Topi
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
| | - Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, Institute of Bari, 70124 Bari, Italy
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
| | - Elona Gaxhja
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
- Doctoral School, eCampus University, 22060 Novedrate, Italy
| |
Collapse
|
13
|
Halpern MD, Gupta A, Zaghloul N, Thulasingam S, Calton CM, Camp SM, Garcia JGN, Ahmed M. Extracellular Nicotinamide Phosphoribosyltransferase Is a Therapeutic Target in Experimental Necrotizing Enterocolitis. Biomedicines 2024; 12:970. [PMID: 38790933 PMCID: PMC11118767 DOI: 10.3390/biomedicines12050970] [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: 02/27/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of prematurity. Postulated mechanisms leading to inflammatory necrosis of the ileum and colon include activation of the pathogen recognition receptor Toll-like receptor 4 (TLR4) and decreased levels of transforming growth factor beta (TGFβ). Extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a novel damage-associated molecular pattern (DAMP), is a TLR4 ligand and plays a role in a number of inflammatory disease processes. To test the hypothesis that eNAMPT is involved in NEC, an eNAMPT-neutralizing monoclonal antibody, ALT-100, was used in a well-established animal model of NEC. Preterm Sprague-Dawley pups delivered prematurely from timed-pregnant dams were exposed to hypoxia/hypothermia and randomized to control-foster mother dam-fed rats, injected IP with saline (vehicle) 48 h after delivery; control + mAB-foster dam-fed rats, injected IP with 10 µg of ALT-100 at 48 h post-delivery; NEC-orally gavaged, formula-fed rats injected with saline; and NEC + mAb-formula-fed rats, injected IP with 10 µg of ALT-100 at 48 h. The distal ileum was processed 96 h after C-section delivery for histological, biochemical, molecular, and RNA sequencing studies. Saline-treated NEC pups exhibited markedly increased fecal blood and histologic ileal damage compared to controls (q < 0.0001), and findings significantly reduced in ALT-100 mAb-treated NEC pups (q < 0.01). Real-time PCR in ileal tissues revealed increased NAMPT in NEC pups compared to pups that received the ALT-100 mAb (p < 0.01). Elevated serum levels of tumor necrosis factor alpha (TNFα), interleukin 6 (IL-6), interleukin-8 (IL-8), and NAMPT were observed in NEC pups compared to NEC + mAb pups (p < 0.01). Finally, RNA-Seq confirmed dysregulated TGFβ and TLR4 signaling pathways in NEC pups that were attenuated by ALT-100 mAb treatment. These data strongly support the involvement of eNAMPT in NEC pathobiology and eNAMPT neutralization as a strategy to address the unmet need for NEC therapeutics.
Collapse
Affiliation(s)
- Melissa D. Halpern
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Senthilkumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Christine M. Calton
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sara M. Camp
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Joe G. N. Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
14
|
van Gorp C, de Lange IH, Hütten MC, López-Iglesias C, Massy KRI, Kessels L, Knoops K, Cuijpers I, Sthijns MMJPE, Troost FJ, van Gemert WG, Spiller OB, Birchenough GMH, Zimmermann LJI, Wolfs TGAM. Antenatal Ureaplasma Infection Causes Colonic Mucus Barrier Defects: Implications for Intestinal Pathologies. Int J Mol Sci 2024; 25:4000. [PMID: 38612809 PMCID: PMC11011967 DOI: 10.3390/ijms25074000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Chorioamnionitis is a risk factor for necrotizing enterocolitis (NEC). Ureaplasma parvum (UP) is clinically the most isolated microorganism in chorioamnionitis, but its pathogenicity remains debated. Chorioamnionitis is associated with ileal barrier changes, but colonic barrier alterations, including those of the mucus barrier, remain under-investigated, despite their importance in NEC pathophysiology. Therefore, in this study, the hypothesis that antenatal UP exposure disturbs colonic mucus barrier integrity, thereby potentially contributing to NEC pathogenesis, was investigated. In an established ovine chorioamnionitis model, lambs were intra-amniotically exposed to UP or saline for 7 d from 122 to 129 d gestational age. Thereafter, colonic mucus layer thickness and functional integrity, underlying mechanisms, including endoplasmic reticulum (ER) stress and redox status, and cellular morphology by transmission electron microscopy were studied. The clinical significance of the experimental findings was verified by examining colon samples from NEC patients and controls. UP-exposed lambs have a thicker but dysfunctional colonic mucus layer in which bacteria-sized beads reach the intestinal epithelium, indicating undesired bacterial contact with the epithelium. This is paralleled by disturbed goblet cell MUC2 folding, pro-apoptotic ER stress and signs of mitochondrial dysfunction in the colonic epithelium. Importantly, the colonic epithelium from human NEC patients showed comparable mitochondrial aberrations, indicating that NEC-associated intestinal barrier injury already occurs during chorioamnionitis. This study underlines the pathogenic potential of UP during pregnancy; it demonstrates that antenatal UP infection leads to severe colonic mucus barrier deficits, providing a mechanistic link between antenatal infections and postnatal NEC development.
Collapse
Affiliation(s)
- Charlotte van Gorp
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Ilse H. de Lange
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Matthias C. Hütten
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Neonatology, Department of Pediatrics, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Kimberly R. I. Massy
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Lilian Kessels
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Kèvin Knoops
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Iris Cuijpers
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Mireille M. J. P. E. Sthijns
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Freddy J. Troost
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Wim G. van Gemert
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Owen B. Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK;
| | - George M. H. Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Luc J. I. Zimmermann
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
15
|
Belei O, Basaca DG, Olariu L, Pantea M, Bozgan D, Nanu A, Sîrbu I, Mărginean O, Enătescu I. The Interaction between Stress and Inflammatory Bowel Disease in Pediatric and Adult Patients. J Clin Med 2024; 13:1361. [PMID: 38592680 PMCID: PMC10932475 DOI: 10.3390/jcm13051361] [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: 01/27/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Inflammatory bowel diseases (IBDs) have seen an exponential increase in incidence, particularly among pediatric patients. Psychological stress is a significant risk factor influencing the disease course. This review assesses the interaction between stress and disease progression, focusing on articles that quantified inflammatory markers in IBD patients exposed to varying degrees of psychological stress. Methods: A systematic narrative literature review was conducted, focusing on the interaction between IBD and stress among adult and pediatric patients, as well as animal subjects. The research involved searching PubMed, Scopus, Medline, and Cochrane Library databases from 2000 to December 2023. Results: The interplay between the intestinal immunity response, the nervous system, and psychological disorders, known as the gut-brain axis, plays a major role in IBD pathophysiology. Various types of stressors alter gut mucosal integrity through different pathways, increasing gut mucosa permeability and promoting bacterial translocation. A denser microbial load in the gut wall emphasizes cytokine production, worsening the disease course. The risk of developing depression and anxiety is higher in IBD patients compared with the general population, and stress is a significant trigger for inducing acute flares of the disease. Conclusions: Further large studies should be conducted to assess the relationship between stressors, psychological disorders, and their impact on the course of IBD. Clinicians involved in the medical care of IBD patients should aim to implement stress reduction practices in addition to pharmacological therapies.
Collapse
Affiliation(s)
- Oana Belei
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (O.M.)
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Diana-Georgiana Basaca
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (O.M.)
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Laura Olariu
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Manuela Pantea
- Twelfth Department, Neonatology Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (M.P.); (I.E.)
| | - Daiana Bozgan
- Clinic of Neonatology, “Pius Brânzeu” County Emergency Clinical Hospital, 300723 Timișoara, Romania;
| | - Anda Nanu
- Third Pediatric Clinic, “Louis Țurcanu” Emergency Children Hospital, 300011 Timișoara, Romania; (A.N.); (I.S.)
| | - Iuliana Sîrbu
- Third Pediatric Clinic, “Louis Țurcanu” Emergency Children Hospital, 300011 Timișoara, Romania; (A.N.); (I.S.)
| | - Otilia Mărginean
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (O.M.)
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Ileana Enătescu
- Twelfth Department, Neonatology Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (M.P.); (I.E.)
| |
Collapse
|
16
|
Stylianou-Riga P, Boutsikou T, Kouis P, Michailidou K, Kinni P, Sokou R, Iliodromiti Z, Pitsios C, Yiallouros PK, Iacovidou N. Epidemiology, risk factors, clinical presentation and complications of late-onset neonatal sepsis among preterm neonates in Cyprus: a prospective case-control study. BMC Pediatr 2024; 24:50. [PMID: 38229029 DOI: 10.1186/s12887-023-04359-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 10/12/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Late-onset neonatal sepsis (LOS) is common in preterm neonates, with increasing incidence in recent years. In the present study, we examined the epidemiology, clinical presentation, and complications of LOS in Cyprus and quantified possible risk factors for the development of this condition. METHODS The study subjects were preterm neonates admitted in the Neonatal Intensive Care Unit (NICU) of Archbishop Makarios III Hospital, the only neonatal tertiary centre in Cyprus. A prospective, case-control study was designed, and carried out between April 2017-October 2018. Depending on blood culture results, preterm neonates were classified as "Confirmed LOS": positive blood culture - microorganism isolated and LOS symptoms, "Unconfirmed LOS": negative blood culture and LOS symptoms, and "Controls" group: negative blood culture and absence of LOS symptoms. Comparisons between the 3 groups were performed and the associations between demographic, clinical and treatment characteristics with the likelihood of LOS were assessed using univariate and multivariate logistic regression. RESULTS A total of 350 preterm neonates were included in the study and the incidence of LOS was 41.1%. 79 (22.6%) and 65 (18.6%) neonates were classified as "Confirmed LOS", and "unconfirmed LOS" cases respectively while 206 (58.9%) served as controls. The rate of confirmed LOS ranged from 12.2% in moderate to late preterm neonates to 78.6% in extremely preterm neonates. In the multivariate model, we demonstrated an independent association between LOS and duration of hospitalization (OR: 1.06, 95%CI: 1.01-1.10), duration of ventilation (OR: 1.23, 95%CI: 1.07-1.43) and necrotising enterocolitis (OR: 3.41, 95%CI: 1.13-10.25). CONCLUSIONS The present study highlights the epidemiology of LOS in preterm neonates in Cyprus and its association with the duration of ventilation and hospitalization as well as with necrotizing enterocolitis. Establishment of protocols for the prevention of nosocomial infections during hospitalization in the NICUs and mechanical ventilation of preterm neonates is recommended.
Collapse
Affiliation(s)
- Paraskevi Stylianou-Riga
- Neonatal Intensive Care Unit, "Archbishop Makarios III" Hospital, Nicosia, Cyprus.
- Respiratory Physiology Laboratory, Medical School, University of Cyprus Shakolas Educational Center of Clinical, Medicine Palaios Dromos Lefkosias Lemesou 215/6, Aglantzia, Nicosia, 2029, Cyprus.
- Neonatal Department, Medical School, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Theodora Boutsikou
- Neonatal Department, Medical School, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panayiotis Kouis
- Respiratory Physiology Laboratory, Medical School, University of Cyprus Shakolas Educational Center of Clinical, Medicine Palaios Dromos Lefkosias Lemesou 215/6, Aglantzia, Nicosia, 2029, Cyprus
| | | | - Paraskevi Kinni
- Respiratory Physiology Laboratory, Medical School, University of Cyprus Shakolas Educational Center of Clinical, Medicine Palaios Dromos Lefkosias Lemesou 215/6, Aglantzia, Nicosia, 2029, Cyprus
| | - Rozeta Sokou
- Neonatal Department, Medical School, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Zoi Iliodromiti
- Neonatal Department, Medical School, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Pitsios
- Respiratory Physiology Laboratory, Medical School, University of Cyprus Shakolas Educational Center of Clinical, Medicine Palaios Dromos Lefkosias Lemesou 215/6, Aglantzia, Nicosia, 2029, Cyprus
| | - Panayiotis K Yiallouros
- Respiratory Physiology Laboratory, Medical School, University of Cyprus Shakolas Educational Center of Clinical, Medicine Palaios Dromos Lefkosias Lemesou 215/6, Aglantzia, Nicosia, 2029, Cyprus
| | - Nicoletta Iacovidou
- Neonatal Department, Medical School, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Leach ST. Role of Probiotics and Prebiotics in Gut Symbiosis. Nutrients 2024; 16:238. [PMID: 38257131 PMCID: PMC10819279 DOI: 10.3390/nu16020238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 01/24/2024] Open
Abstract
The objective of this Special Issue entitled 'Role of Probiotics and Prebiotics in Gut Symbiosis' is to publish reviews, clinical trials and experimental studies that focus on probiotics and prebiotics that have a role in influencing disease and promoting gastrointestinal and overall health [...].
Collapse
Affiliation(s)
- Steven T Leach
- Department of Paediatrics, School of Clinical Medicine, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
18
|
Calton CM, Carothers K, Ramamurthy S, Jagadish N, Phanindra B, Garcia A, Viswanathan VK, Halpern MD. Clostridium scindens exacerbates experimental necrotizing enterocolitis via upregulation of the apical sodium-dependent bile acid transporter. Am J Physiol Gastrointest Liver Physiol 2024; 326:G25-G37. [PMID: 37933481 PMCID: PMC11208032 DOI: 10.1152/ajpgi.00102.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in premature infants. Evidence indicates that bile acid homeostasis is disrupted during NEC: ileal bile acid levels are elevated in animals with experimental NEC, as is expression of the apical sodium-dependent bile acid transporter (Asbt). In addition, bile acids, which are synthesized in the liver, are extensively modified by the gut microbiome, including via the conversion of primary bile acids to more cytotoxic secondary forms. We hypothesized that the addition of bile acid-modifying bacteria would increase susceptibility to NEC in a neonatal rat model of the disease. The secondary bile acid-producing species Clostridium scindens exacerbated both incidence and severity of NEC. C. scindens upregulated the bile acid transporter Asbt and increased levels of intraenterocyte bile acids. Treatment with C. scindens also altered bile acid profiles and increased hydrophobicity of the ileal intracellular bile acid pool. The ability of C. scindens to enhance NEC requires bile acids, as pharmacological sequestration of ileal bile acids protects animals from developing disease. These findings indicate that bile acid-modifying bacteria can contribute to NEC pathology and provide additional evidence for the role of bile acids in the pathophysiology of experimental NEC.NEW & NOTEWORTHY Necrotizing enterocolitis (NEC), a life-threatening gastrointestinal emergency in premature infants, is characterized by dysregulation of bile acid homeostasis. We demonstrate that administering the secondary bile acid-producing bacterium Clostridium scindens enhances NEC in a neonatal rat model of the disease. C. scindens-enhanced NEC is dependent on bile acids and driven by upregulation of the ileal bile acid transporter Asbt. This is the first report of bile acid-modifying bacteria exacerbating experimental NEC pathology.
Collapse
Affiliation(s)
- Christine M Calton
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - Katelyn Carothers
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States
| | - Shylaja Ramamurthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States
| | - Neha Jagadish
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - Bhumika Phanindra
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - Anett Garcia
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - V K Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States
| | - Melissa D Halpern
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
19
|
Iijima S. Clinical Dilemma Involving Treatments for Very Low-Birth-Weight Infants and the Potential Risk of Necrotizing Enterocolitis: A Narrative Literature Review. J Clin Med 2023; 13:62. [PMID: 38202069 PMCID: PMC10780023 DOI: 10.3390/jcm13010062] [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: 11/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a critical gastrointestinal emergency with substantial morbidity and mortality risks, especially for very low-birth-weight (VLBW) infants, and unclear multifactorial pathophysiology. Whether common treatments for VLBW infants increase the NEC risk remains controversial. Indomethacin (utilized for patent ductus arteriosus) offers benefits but is concerning because of its vasoconstrictive impact on NEC susceptibility. Similarly, corticosteroids used to treat bronchopulmonary dysplasia may increase vulnerability to NEC by compromising immunity and altering the mesenteric blood flow. Histamine-2 receptor blockers (used to treat gastric bleeding) may inadvertently promote NEC by affecting bacterial colonization and translocation. Doxapram (used to treat apnea) poses a risk of gastrointestinal disturbance via gastric acid hypersecretion and circulatory changes. Glycerin enemas aid meconium evacuation but disrupt microbial equilibrium and trigger stress-related effects associated with the NEC risk. Prolonged antibiotic use may unintentionally increase the NEC risk. Blood transfusions for anemia can promote NEC via interactions between the immune response and ischemia-reperfusion injury. Probiotics for NEC prevention are associated with concerns regarding sepsis and bacteremia. Amid conflicting evidence, this review unveils NEC risk factors related to treatments for VLBW infants, offers a comprehensive overview of the current research, and guides personalized management strategies, thereby elucidating this clinical dilemma.
Collapse
Affiliation(s)
- Shigeo Iijima
- Department of Regional Neonatal-Perinatal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| |
Collapse
|
20
|
Huang YC, Hsu KH, Chu SM, Chiang MC, Lien R, Chen KJ, Hwang YS, Lai CC, Tseng HJ, Wu WC. Respiratory outcomes in preterm infants following intravitreal bevacizumab for retinopathy of prematurity-a 10-year matched case study. Eye (Lond) 2023; 37:3675-3681. [PMID: 37400566 PMCID: PMC10686401 DOI: 10.1038/s41433-023-02579-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/05/2023] [Accepted: 05/11/2023] [Indexed: 07/05/2023] Open
Abstract
OBJECTIVES To evaluate respiratory outcomes in preterm infants with retinopathy of prematurity (ROP) following intravitreal bevacizumab injection (IVB). METHODS This single-centre study enroled preterm infants with a gestational age (GA) < 34 weeks or a birth weight (BW) < 1500 g with bilateral type 1 ROP who received a single IVB, and a treatment-free control group matched by GA, postmenstrual age, and respiratory status at the time of the IVB. The primary outcome was serial respiratory changes in mean airway pressure (MAP), fraction of inspired oxygen (FiO2), and respiratory severity score (RSS, MAP x FiO2) during the 28-day post-IVB/matching period and overall respiratory improvement at day 28 and at discharge. The duration of supplemental oxygen therapy following IVB/matching was documented. RESULTS A total of 5578 infants were included. Seventy-eight infants were enroled in the IVB group, and another 78 infants were matched as the control group. Both groups had downward trends in the MAP, FiO2, and RSS over the study period (all P < 0.001), but there were no between-group differences in these measures. The percentage of overall respiratory improvement was similar between the IVB and control groups, so was the duration of invasive and in-hospital oxygen ventilation. A lower percentage of oxygen dependence at discharge in the IVB group (P = 0.03) remained significant after adjusting for GA and BW. CONCLUSIONS This is a matched case study to evaluate respiratory outcomes in preterm infants following IVB for ROP. We found that the IVBs did not compromise respiratory outcomes in preterm infants during the 28-day post-IVB period and at discharge.
Collapse
Affiliation(s)
- Ying-Chen Huang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Kai-Hsiang Hsu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Science, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Ming Chu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Chou Chiang
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Science, Chang Gung University, Taoyuan, Taiwan
| | - Reyin Lien
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuan-Jen Chen
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yih-Shiou Hwang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Chun Lai
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Hsiao-Jung Tseng
- Biostatistics unit, Clinical Trial Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wei-Chi Wu
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
21
|
Gómez-Ferrer M, Amaro-Prellezo E, Albiach-Delgado A, Ten-Domenech I, Kuligowski J, Sepúlveda P. Identification of omega-3 oxylipins in human milk-derived extracellular vesicles with pro-resolutive actions in gastrointestinal inflammation. Front Immunol 2023; 14:1293737. [PMID: 38054009 PMCID: PMC10694275 DOI: 10.3389/fimmu.2023.1293737] [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: 09/13/2023] [Accepted: 10/27/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction Premature infants (PIs) are at risk of suffering necrotizing enterocolitis (NEC), and infants consuming human milk (HM) show a lower incidence than infants receiving formula. The composition of HM has been studied in depth, but the lipid content of HM-derived small extracellular vesicles (HM sEVs) remains unexplored. Identifying these molecules and their biological effects has potential for the treatment of intestinal disorders in PIs and could contribute to the development of HM-based fortified formulas. Methods We isolated HM sEVs from HM samples and analyzed their oxylipin content using liquid chromatography coupled to mass spectrometry, which revealed the presence of anti-inflammatory oxylipins. We then examined the efficacy of a mixture of these oxylipins in combating inflammation and fibrosis, in vitro and in a murine model of inflammatory bowel disease (IBD). Results HM-related sEVs contained higher concentrations of oxylipins derived from docosahexaenoic acid, an omega-3 fatty acid. Three anti-inflammatory oxylipins, 14-HDHA, 17-HDHA, and 19,20-DiHDPA (ω3 OXLP), demonstrated similar efficacy to HM sEVs in preventing cell injury, inducing re-epithelialization, mitigating fibrosis, and modulating immune responses. Both ω3 OXLP and HM sEVs effectively reduced inflammation in IBD-model mice, preventing colon shortening, infiltration of inflammatory cells and tissue fibrosis. Discussion Incorporating this unique cocktail of oxylipins into fortified milk formulas might reduce the risk of NEC in PIs and also provide immunological and neurodevelopmental support.
Collapse
Affiliation(s)
- Marta Gómez-Ferrer
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Elena Amaro-Prellezo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Abel Albiach-Delgado
- Neonatal Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Isabel Ten-Domenech
- Neonatal Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Valencia, Spain
- Cardiology Service, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, Madrid, Spain
- Department of Pathology, University of Valencia, Valencia, Spain
| |
Collapse
|
22
|
Lin X, Wu C. Identification and evaluation of probiotic potential of Bifidobacterium breve AHC3 isolated from chicken intestines and its effect on necrotizing enterocolitis (NEC) in newborn SD rats. PLoS One 2023; 18:e0287799. [PMID: 37917716 PMCID: PMC10621988 DOI: 10.1371/journal.pone.0287799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/13/2023] [Indexed: 11/04/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe intestinal disease of the newborn infants, associated with high morbidity and mortality. It has been reported that Bifidobacterium could protect the intestinal barrier function and reduce the risk of NEC. This study aimed to evaluate the probiotic potential of Bifidobacterium strains isolated from the chicken intestines and its effect on necrotizing enterocolitis in newborn SD rats. Out of 32 isolates, B. breve AHC3 not only exhibited excellent probiotic potential, including tolerance to artificial simulated gastric conditions, adhesion to HT-29 cells, antioxidant capacity and antibacterial activity, but also possessed reliable safety. Additionally, NEC model was established to further investigate the effect of B. breve AHC3 on necrotizing enterocolitis in newborn SD rats. It was illustrated that administration of B. breve AHC3 significantly not only reduced the incidence of NEC (from 81.25% to 34.38%) (P< 0.05), but also alleviated the severity of ileal injury (P< 0.05). Compared with NEC model, B. breve AHC3 could significantly decrease the level of proinflammatory factor TNF-α (P< 0.05) and increase the level of antiinflammatory factor IL-10 (P< 0.05) in the ileum of NEC rats. Through the intervention of B. breve AHC3, the gray value of inducible nitric oxide synthase (iNOS) in intestinal tissue of NEC rats was significantly reduced (P< 0.05). It was indicated that B. breve AHC3 exhibited prominent probiotic potential and reliable safety. In the neonatal SD rat model of NEC, B. breve AHC3 had an available protective effect on the intestinal injury of NEC, which might be related to reducing the inflammatory reaction in the ileum and inhibiting the expression of iNOS in intestinal tissue cells. B. breve AHC3 could be used as a potential treatment for human NEC.
Collapse
Affiliation(s)
- Xiaopei Lin
- Department of Pediatrics, Maternity and Child Health Care Hospital Affiliated to Anhui Medical University (Anhui Maternity and Child Health Care Hospital), Hefei, Anhui, China
| | - Changjun Wu
- Institute of Microbiology, Anhui Academy of Medical Sciences, Hefei, Anhui, China
| |
Collapse
|
23
|
Zaikova EK, Kaplina AV, Petrova NA, Pervunina TM, Kostareva AA, Kalinina OV. SIGIRR gene variants in term newborns with congenital heart defects and necrotizing enterocolitis. Ann Pediatr Cardiol 2023; 16:337-344. [PMID: 38766461 PMCID: PMC11098289 DOI: 10.4103/apc.apc_30_23] [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: 03/01/2023] [Revised: 03/28/2023] [Accepted: 08/03/2023] [Indexed: 05/22/2024] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a common gastrointestinal emergency among neonates which is characterized by acute intestinal inflammation and necrosis. The main risk factors for NEC are prematurity, low birth weight, and some preexisting health conditions such as congenital heart defects (CHDs). Investigation of the potential genetic predisposition to NEC is a promising approach that might provide new insights into its pathogenesis. One of the most important proteins that play a significant role in the pathogenesis of NEC is Toll-like receptor 4 (TLR4) which recognizes lipopolysaccharide found in Gram-negative bacteria. In intestinal epithelial cells, a protein encoded by the SIGIRR gene is a major inhibitor of TLR4 signaling. A few SIGIRR variants, including rare p.Y168X and p.S80Y, have already been identified in preterm infants with NEC, but their pathogenic significance remains unclear. This study aimed to investigate the spectrum of SIGIRR genetic variants in term newborns with CHD and to assess their potential association with NEC. Methods and Results A total of 93 term newborns with critical CHD were enrolled in this study, 33 of them developed NEC. SIGIRR genetic variants were determined by Sanger sequencing of all exons. In total, eight SIGIRR genetic variants were identified, two of which were found only in newborns with NEC (P = 0.12). The rare missense p.S80Y (rs117739035) variant in exon 4 was found in two infants with NEC stage IIA. Two infants with NEC stage III and stage IB carried a novel duplication c. 102_121dup (rs552367848) variant in exon 10 that has not been previously associated with any clinical phenotype. Conclusions The presence of both variants only in neonates who developed NEC, together with earlier published data, may suggest their potential contribution to the risk of developing NEC in term infants with CHD and allow planning larger cohort studies to clarify their relevance.
Collapse
Affiliation(s)
- Ekaterina Konstantinovna Zaikova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Research Laboratory of Autoimmune and Autoinflammatory Diseases, St. Petersburg, Russia
| | - Aleksandra Vladimirovna Kaplina
- Almazov National Medical Research Centre, Research Laboratory of Physiology and Diseases of Newborns, St. Petersburg, Russia
| | - Natalia Aleksandrovna Petrova
- Almazov National Medical Research Centre, Research Laboratory of Physiology and Diseases of Newborns, St. Petersburg, Russia
| | | | | | - Olga Viktorovna Kalinina
- Almazov National Medical Research Centre, Research Laboratory of Physiology and Diseases of Newborns, St. Petersburg, Russia
- Department of Laboratory Medicine and Genetics, Institution of Medical Education, Almazov National Medical Research Centre, St. Petersburg, Russia
| |
Collapse
|
24
|
Men G, Wang L, Lu X, Wen G, Lü Q. Can Enterococcus faecium prevent NEC in preterm infants?: A systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e34787. [PMID: 37565851 PMCID: PMC10419755 DOI: 10.1097/md.0000000000034787] [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] [Received: 06/01/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE Previous some systematic reviews reported that probiotics may benefit the prevention of NEC in preterm infants. But dissimilar bacterial strains and taxa used in included studies possibly result in bias. There is not a rounded systematic review which has estimated the benefit and safety of Enterococcus faecium to prevent NEC in preterm infants to date before we conducted. METHODS This systematic review of randomized controlled trials and retrospective studies analyzing the benefit of Enterococcus faecium to prevent NEC in preterm infants was performed using PubMed, Web of Science, Cochrane Library, EMBASE, Wanfang data and China National Knowledge Infrastructure databases from inception to April 14, 2023. The search terms were "preterm" AND "necrotizing enterocolitis" AND "Enterococcus faecium OR probiotics." Studies reporting NEC involving preterm infants who were given Enterococcus faecium were included in this systematic review. A sensitivity analysis was conducted to assess the stability of results. A funnel plot was generated to identify publication bias. Two authors appraised studies quality and extracted data independently. This work has been reported according with preferred reporting items for systematic reviews and meta-analyses and assessing the methodological quality of systematic reviews. Statistical analysis was conducted using Review Manager 5.3 software. Risk ratio (RR) with 95% confidence intervals (CI) was calculated and analyzed. RESULTS Seven studies (N = 1487 participants) were included in this systematic review, and 6 randomized, controlled trials (N = 1237 participants) were included in the meta-analysis. Comparing with the control groups, the Enterococcus faecium groups had a significant decline in the incidence of NEC Bell stage II or higher (RR: 0.3138, 95% CI: 0.1983-0.4965; P < .00001; 6 studies, n = 1237) and infection (RR: 0.4818, 95% CI: 0.2950-0.7869; P = .004; 3 studies, n = 710). CONCLUSIONS Enterococcus faecium is effective and safe in preventing NEC (Bell stage II or higher) in preterm infants. But all studies included came from China. The dosages and durations of taking Enterococcus faecium were various.
Collapse
Affiliation(s)
- Guangguo Men
- Neonatal Intensive Care Unit, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Lili Wang
- Department of Neonatology, Dong’e Hospital of Traditional Chinese Medicine, Liaocheng, Shandong, China
| | - Xudan Lu
- Department of Neonatology, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Gang Wen
- Department of Pediatric Surgery, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Qin Lü
- Neonatal Intensive Care Unit, Ningbo Women and Children’s Hospital, Ningbo, China
| |
Collapse
|
25
|
Menchaca AD, Style CC, Kyhl TA, Chawla M, Texter KM, Olutoye OO. Inhaled Nitric Oxide and Higher Necrotizing Enterocolitis Rates in Congenital Heart Disease Patients. J Surg Res 2023; 288:166-171. [PMID: 36989832 DOI: 10.1016/j.jss.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/12/2023] [Accepted: 02/18/2023] [Indexed: 03/29/2023]
Abstract
INTRODUCTION Certain congenital cardiac lesions are at increased risk for the development of necrotizing enterocolitis (NEC). These patients are often reliant on pulmonary and systemic vasomodulators to maintain adequate perfusion and oxygenation. This study sought to determine whether pulmonary or systemic vasodilator treatment is protective against the development of NEC in this population. METHODS We utilized International Classification of Diseases (ICD) codes to identify high risk congenital cardiac disease patients ≤6 mo of age, cared for at a tertiary children's hospital between January 2011 and January 2021. Cardiac anomalies were stratified into ductal dependent (pulmonary DD-P or systemic DD-S) or independent lesions. The rate of NEC development in those who received vasodilators (inhaled nitric oxide [iNO], pulmonary vasodilators, systemic vasodilators) was compared to controls in a multivariate analysis. RESULTS Of the 352 patients, who met inclusion criteria, 77.6% had ductal dependent lesions (DD-S 41.9%, DD-P 35.7%), 19.5% received iNO, and 37.5% received other vasodilatory drugs. The overall NEC rate was 15.1%. On univariate analysis, DD-S, iNO use, and systemic vasodilators was associated with a significantly higher risk of NEC, while DD-P was associated with lower NEC risk. On multivariate analysis, only iNO (odds ratio 2.725, confidence interval [1.36-5.44]) and DD-S (odds ratio 2.279, confidence interval [1.02-5.11]) were independent risk factors for NEC. CONCLUSIONS In patients with at-risk congenital cardiac disease lesions, a ductus dependent systemic circulation or iNO treatment is associated with an increased risk of developing NEC. The presence of iNO or DD-S should be utilized as markers of increased risk both in the prevention and workup of suspected NEC.
Collapse
|
26
|
Gephart SM, Fleiner M, Msowoya A, Rothers J. Prediction of GutCheck NEC and Its Relation to Severity of Illness and Measures of Deterioration in Necrotizing Enterocolitis. Adv Neonatal Care 2023; 23:377-386. [PMID: 37339581 PMCID: PMC10440277 DOI: 10.1097/anc.0000000000001080] [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] [Indexed: 06/22/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) risk has been shown to arise from multiple sources and risk awareness may be supported using bedside tools. PURPOSE The purpose of this research was to examine the extent to which GutCheck NEC was associated with scores for clinical deterioration, severity of illness, and clinical outcome, and further to examine how scores might improve NEC prediction. METHODS A retrospective, correlational case-control study with infant data from 3 affiliated neonatal intensive care units was conducted. RESULTS Of 132 infants (44 cases, 88 controls), most were 28 weeks of gestation at birth and less (74%). Median age at NEC onset was 18 days (range: 6-34 days), with two-thirds diagnosed before 21 days. At 68 hours of life, higher GutCheck NEC scores were associated with NEC requiring surgery or resulting in death (relative risk ratio [RRR] = 1.06, P = .036), associations that persisted at 24 hours prior to diagnosis (RRR = 1.05, P = .046), and at the time of diagnosis (RRR = 1.05, P = .022) but showed no associations for medical NEC. GutCheck NEC scores were significantly correlated with pediatric early warning scores (PEWS) ( r > 0.30; P < .005) and SNAPPE-II scores ( r > 0.44, P < .0001). Increasing numbers of clinical signs and symptoms were positively associated with GutCheck NEC and PEWS at the time of diagnosis ( r = 0.19, P = .026; and r = 0.25, P = .005, respectively). IMPLICATIONS FOR PRACTICE AND RESEARCH GutCheck NEC provides structure to streamline assessment and communication about NEC risk. Yet, it is not intended to be diagnostic. Research is needed on how GutCheck NEC impacts timely recognition and treatment.
Collapse
Affiliation(s)
- Sheila M Gephart
- College of Nursing, The University of Arizona, Tucson (Drs Gephart and Rothers); Neonatal Intensive Care Clinical Nurse Specialist, Banner Health, Mesa, Arizona (Dr Fleiner); Karibu Family Care, Peoria, Arizona (Dr Msowoya); and StatLab BIO5 Institute, The University of Arizona, Tucson (Dr Rothers)
| | | | | | | |
Collapse
|
27
|
McElroy SJ, Lueschow SR. State of the art review on machine learning and artificial intelligence in the study of neonatal necrotizing enterocolitis. Front Pediatr 2023; 11:1182597. [PMID: 37303753 PMCID: PMC10250644 DOI: 10.3389/fped.2023.1182597] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/25/2023] [Indexed: 06/13/2023] Open
Abstract
Necrotizing Enterocolitis (NEC) is one of the leading causes of gastrointestinal emergency in preterm infants. Although NEC was formally described in the 1960's, there is still difficulty in diagnosis and ultimately treatment for NEC due in part to the multifactorial nature of the disease. Artificial intelligence (AI) and machine learning (ML) techniques have been applied by healthcare researchers over the past 30 years to better understand various diseases. Specifically, NEC researchers have used AI and ML to predict NEC diagnosis, NEC prognosis, discover biomarkers, and evaluate treatment strategies. In this review, we discuss AI and ML techniques, the current literature that has applied AI and ML to NEC, and some of the limitations in the field.
Collapse
Affiliation(s)
- Steven J. McElroy
- Department of Pediatrics, University of California Davis, Sacramento, CA, United States
| | - Shiloh R. Lueschow
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
28
|
Ragan MV, Wala SJ, Sajankila N, Duff AF, Wang Y, Volpe SG, Al-Hadidi A, Dumbauld Z, Purayil N, Wickham J, Conces MR, Mihi B, Goodman SD, Bailey MT, Besner GE. Development of a novel definitive scoring system for an enteral feed-only model of necrotizing enterocolitis in piglets. Front Pediatr 2023; 11:1126552. [PMID: 37138566 PMCID: PMC10149862 DOI: 10.3389/fped.2023.1126552] [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: 12/18/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Necrotizing enterocolitis (NEC) is a complex inflammatory disorder of the human intestine that most often occurs in premature newborns. Animal models of NEC typically use mice or rats; however, pigs have emerged as a viable alternative given their similar size, intestinal development, and physiology compared to humans. While most piglet NEC models initially administer total parenteral nutrition prior to enteral feeds, here we describe an enteral-feed only piglet model of NEC that recapitulates the microbiome abnormalities present in neonates that develop NEC and introduce a novel multifactorial definitive NEC (D-NEC) scoring system to assess disease severity. Methods Premature piglets were delivered via Caesarean section. Piglets in the colostrum-fed group received bovine colostrum feeds only throughout the experiment. Piglets in the formula-fed group received colostrum for the first 24 h of life, followed by Neocate Junior to induce intestinal injury. The presence of at least 3 of the following 4 criteria were required to diagnose D-NEC: (1) gross injury score ≥4 of 6; (2) histologic injury score ≥3 of 5; (3) a newly developed clinical sickness score ≥5 of 8 within the last 12 h of life; and (4) bacterial translocation to ≥2 internal organs. Quantitative reverse transcription polymerase chain reaction was performed to confirm intestinal inflammation in the small intestine and colon. 16S rRNA sequencing was performed to evaluate the intestinal microbiome. Results Compared to the colostrum-fed group, the formula-fed group had lower survival, higher clinical sickness scores, and more severe gross and histologic intestinal injury. There was significantly increased bacterial translocation, D-NEC, and expression of IL-1α and IL-10 in the colon of formula-fed compared to colostrum-fed piglets. Intestinal microbiome analysis of piglets with D-NEC demonstrated lower microbial diversity and increased Gammaproteobacteria and Enterobacteriaceae. Conclusions We have developed a clinical sickness score and a new multifactorial D-NEC scoring system to accurately evaluate an enteral feed-only piglet model of NEC. Piglets with D-NEC had microbiome changes consistent with those seen in preterm infants with NEC. This model can be used to test future novel therapies to treat and prevent this devastating disease.
Collapse
Affiliation(s)
- Mecklin V. Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Samantha J. Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Nitin Sajankila
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Audrey F. Duff
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Yijie Wang
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Samuel G. Volpe
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Ameer Al-Hadidi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Zachary Dumbauld
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Nanditha Purayil
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Joseph Wickham
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Miriam R. Conces
- Department of Pathology, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Belgacem Mihi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Michael T. Bailey
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Gail E. Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| |
Collapse
|
29
|
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of death and disability from gastrointestinal disease in premature infants. The mortality of patients with NEC is approximately 30%, a figure that has not changed in many decades, reflecting the need for a greater understanding of its pathogenesis. Progress towards understanding the cellular and molecular mechanisms underlying NEC requires the study of highly translational animal models. Such animal models must mimic the biology and physiology of premature infants, while still allowing for safe experimental manipulation of environmental and microbial factors thought to be associated with the risk and severity of NEC. Findings from animal models have yielded insights into the interactions between the host, the colonizing microbes, and the innate immune receptor Toll-like Receptor 4 (TLR4) in driving disease development. This review discusses the relative strengths and weaknesses of available in vivo, in vitro, and NEC-in-a-dish models of this disease. We also highlight the unique contributions that each model has made to our understanding of the complex interactions between enterocytes, microbiota, and immune cells in the pathogenesis of NEC. The overall purpose of this review is to provide a menu of options regarding currently available animal models of NEC, while in parallel hopefully reducing the potential uncertainty and confusion regarding NEC models to assist those who wish to enter this field from other disciplines.
Collapse
Affiliation(s)
- Carla M Lopez
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Maame Efua S Sampah
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Johannes W Duess
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Asuka Ishiyama
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Raheel Ahmad
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Chhinder P Sodhi
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - David J Hackam
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA.
| |
Collapse
|
30
|
Tomo CK, Balogun OO, Davidson J, Guinsburg R, Almeida MFBD, Lopes JMDA, Barros MCDM, Takehara K, Mikami M, Isayama T, Hoshino A, Mori R, Mizuguchi M. Comparison of mortality and survival without major morbidities of very preterm infants with very low birth weight from Japan and Brazil. REVISTA PAULISTA DE PEDIATRIA 2023; 41:e2021389. [PMID: 36102406 PMCID: PMC9462411 DOI: 10.1590/1984-0462/2023/41/2021389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/13/2022] [Indexed: 11/22/2022]
Abstract
Objective: This study was carried out to understand the disparities in mortality and survival without major morbidities among very premature and very low birth weight infants between participating Neonatal Intensive Care Units (NICUs) from the Brazilian Network on Neonatal Research (RBPN) and the Neonatal Research Network of Japan (NRNJ). Methods: Secondary data analysis of surveys by the RBPN and NRNJ was performed. The surveys were conducted in 2014 and 2015 and included 187 NICUs. Primary outcome was mortality or survival without any major morbidity. Logistic regression analysis adjustment for confounding factors was used. Results: The study population consisted of 6,406 infants from the NRNJ and 2,319 from the RBPN. Controlling for various confounders, infants from RBPN had 9.06 times higher adjusted odds of mortality (95%CI 7.30–11.29), and lower odds of survival without major morbidities (AOR 0.36; 95%CI 0.32–0.41) compared with those from the NRNJ. Factors associated with higher odds of mortality among Brazilian NICUs included: Air Leak Syndrome (AOR 4.73; 95%CI 1.26–15.27), Necrotizing Enterocolitis (AOR 3.25; 95%CI 1.38–7.26), and Late Onset Sepsis (LOS) (AOR 4.86; 95%CI 2.25–10.97). Conclusions: Very premature and very low birth weight infants from Brazil had significantly higher odds for mortality and lower odds for survival without major morbidities in comparison to those from Japan. Additionally, we identified the factors that increased the odds of in-hospital neonatal death in Brazil, most of which was related to LOS.
Collapse
Affiliation(s)
- Caroline Kaori Tomo
- University of Tokyo, Japan; National Center for Child Health and Development, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Pugh CP, Baber M, White G. Necrotizing enterocolitis following treatment of congenital syphilis with penicillin in a term newborn. SAGE Open Med Case Rep 2023; 11:2050313X231172672. [PMID: 37205159 PMCID: PMC10186574 DOI: 10.1177/2050313x231172672] [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: 11/29/2022] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Necrotizing enterocolitis is a disease process of intestinal disruption which has been associated with gastrointestinal microbial alterations after antibiotic exposure. Treatment guidelines and antibiotic exposure for congenital syphilis have historically been based on limited evidence. This case presents a term infant who developed necrotizing enterocolitis after treatment for congenital syphilis.
Collapse
Affiliation(s)
- Charles Preston Pugh
- Charles Preston Pugh, Department of Pediatrics,
University of Arkansas for Medical Sciences, 4301 W. Markham Street, Slot 512-5B, Little
Rock, AR 72205, USA.
| | | | | |
Collapse
|
32
|
Yan X, Liu L, Yao S, Chen Y, Yu Q, Jiang C, Chen W, Chen X, Han S. LncRNA and mRNA profiles of human milk-derived exosomes and their possible roles in protecting against necrotizing enterocolitis. Food Funct 2022; 13:12953-12965. [PMID: 36448375 DOI: 10.1039/d2fo01866g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Necrotizing enterocolitis (NEC) is one of the most severe diseases commonly afflicting premature infants. Our previous studies suggests that human milk-derived exosomes (HM-Exos) have a potential therapeutic effect on NEC. In this study, we investigate the potentially therapeutic role of HM-Exos in an NEC animal model via comprehensive lncRNA and mRNA expression profiles. A rat model of NEC was induced through hypoxia, hypothermia and formula feeds. We extracted exosomes from the colostrum of healthy lactating mothers and identified their functions in an NEC animal model. Furthermore, high-throughput lncRNA and mRNA sequencings were explored to find the underlying mechanisms. Although both exosomes from term human breast milk (Term-Exos) and exosomes from preterm human breast milk (Pre-Exos) alleviated the severity of NEC, Pre-Exos seemed to better promote the proliferation of intestinal epithelial cells in vivo. We identified a total of 44 differentially expressed lncRNAs and 88 differentially expressed mRNAs between Term-Exos and Pre-Exos. Further GO and KEGG pathway analysis showed that the lncRNA-mRNA network of HM-Exos was associated with the JAK-STAT signaling pathway, bile secretion and the AMPK signaling pathway, which are predicted to be involved in the proliferation of cells. Therefore, this study reveals for the first time the important roles of human milk derived lncRNAs and mRNAs in protecting against necrotizing enterocolitis. These results provide new insight into the development of NEC.
Collapse
Affiliation(s)
- Xiangyun Yan
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Linjie Liu
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Shuwen Yao
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Yanjie Chen
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Qinlei Yu
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Chengyao Jiang
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Wenjuan Chen
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Xiaohui Chen
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Shuping Han
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| |
Collapse
|
33
|
Machine learning-based risk factor analysis of necrotizing enterocolitis in very low birth weight infants. Sci Rep 2022; 12:21407. [PMID: 36496465 PMCID: PMC9741654 DOI: 10.1038/s41598-022-25746-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
This study used machine learning and a national prospective cohort registry database to analyze the major risk factors of necrotizing enterocolitis (NEC) in very low birth weight (VLBW) infants, including environmental factors. The data consisted of 10,353 VLBW infants from the Korean Neonatal Network database from January 2013 to December 2017. The dependent variable was NEC. Seventy-four predictors, including ambient temperature and particulate matter, were included. An artificial neural network, decision tree, logistic regression, naïve Bayes, random forest, and support vector machine were used to evaluate the major predictors of NEC. Among the six prediction models, logistic regression and random forest had the best performance (accuracy: 0.93 and 0.93, area under the receiver-operating-characteristic curve: 0.73 and 0.72, respectively). According to random forest variable importance, major predictors of NEC were birth weight, birth weight Z-score, maternal age, gestational age, average birth year temperature, birth year, minimum birth year temperature, maximum birth year temperature, sepsis, and male sex. To the best of our knowledge, the performance of random forest in this study was among the highest in this line of research. NEC is strongly associated with ambient birth year temperature, as well as maternal and neonatal predictors.
Collapse
|
34
|
Karvonen KL, Goronga F, McKenzie-Sampson S, Rogers EE. Racial disparities in the development of comorbid conditions after preterm birth: A narrative review. Semin Perinatol 2022; 46:151657. [PMID: 36153273 DOI: 10.1016/j.semperi.2022.151657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite recognition and attempts to reduce racial disparities in perinatal outcomes, Black infants are still disproportionately represented among those who are born preterm. Postnatal investigations of racial disparities in comorbidities and outcomes after preterm birth are increasing, although their results and interpretations are conflicting. In the present review, we 1.) identify important methodological limitations of that literature 2.) summarize the conflicting literature investigating racial disparities, specifically Black-white differences, in postnatal comorbidities and outcomes after preterm birth 3.) describe mechanisms by which racism operates to contextualize our understanding to inform future work to actively reduce disparities in preterm birth and subsequently, its complications.
Collapse
Affiliation(s)
- Kayla L Karvonen
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, United States.
| | - Faith Goronga
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
| | - Safyer McKenzie-Sampson
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, United States; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Elizabeth E Rogers
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
35
|
Shen R, Embleton N, Lyng Forman J, Gale C, Griesen G, Sangild PT, Uthaya S, Berrington J. Early antibiotic use and incidence of necrotising enterocolitis in very preterm infants: a protocol for a UK based observational study using routinely recorded data. BMJ Open 2022; 12:e065934. [PMID: 36379645 PMCID: PMC9667987 DOI: 10.1136/bmjopen-2022-065934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Necrotising enterocolitis (NEC) remains a major contributor to preterm mortality and morbidity. Prolonged duration of antibiotic therapy after delivery is associated with later NEC development but recent evidence suggests that absence of antibiotic treatment after delivery may also increase NEC risk. We will explore this controversy using a large pre-existing dataset of preterm infants in the UK. METHODS AND ANALYSIS This is a retrospective cohort study using data from UK National Neonatal Research Database (NNRD) for infants born 1 January 2012 to 31 December 2020. Eligible infants will be <32 weeks gestation, alive on day 3. Primary outcome is development of severe NEC, compared in infants receiving early antibiotics (days 1-2 after birth) and those not. Subgroup analysis on duration of early antibiotic exposure will also occur. Secondary outcomes are: late onset sepsis, total antibiotic use, predischarge mortality, retinopathy of prematurity, intraventricular haemorrhage, bronchopulmonary dysplasia, focal intestinal perforation and any abdominal surgery. To address competing risks, incidence of death before day 7, 14 and 28 will be analysed. We will perform logistic regression and propensity score matched analyses. Statistical analyses will be guided by NEC risk factors, exposures and outcome presented in a causal diagram. These covariates include but are not limited to gestational age, birth weight, small for gestational age, sex, ethnicity, delivery mode, delivery without labour, Apgar score, early feeding and probiotic use. Sensitivity analyses of alternate NEC definitions, specific antibiotics and time of initiation will occur. ETHICS AND DISSEMINATION We will use deidentified data from NNRD, which holds permissions for the original data, from which parents can opt out and seek study-specific research ethics approval. The results will help to determine optimal use of early antibiotics for very preterm infants. IMPLICATIONS This data will help optimise early antibiotic use in preterm infants. TRIAL REGISTRATION NUMBER ISRCTN55101779.
Collapse
Affiliation(s)
- Rene Shen
- Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhgaen, Denmark
| | - Nicholas Embleton
- Neonatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Population Health Sciences, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | | | - Chris Gale
- Neonatal Medicine, School of Public Health, Imperial College London, London, UK
| | - Gorm Griesen
- Neonatology, Odense University Hospital, Odense, Denmark
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhgaen, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Sabita Uthaya
- Faculty of Medicine, Imperial College London, London, UK
| | - Janet Berrington
- Neonatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Clinical and Translational Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
| |
Collapse
|
36
|
Varisco G, Peng Z, Kommers D, Zhan Z, Cottaar W, Andriessen P, Long X, van Pul C. Central apnea detection in premature infants using machine learning. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 226:107155. [PMID: 36215858 DOI: 10.1016/j.cmpb.2022.107155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND OBJECTIVE Apnea of prematurity is one of the most common diagnosis in neonatal intensive care units. Apneas can be classified as central, obstructive or mixed. According to the current international standards, minimal fluctuations or absence of fluctuations in the chest impedance (CI) suggest a central apnea (CA). However, automatic detection of reduced CI fluctuations leads to a high number of central apnea-suspected events (CASEs), the majority being false alarms. We aim to improve automatic detection of CAs by using machine learning to optimize detection of CAs among CASEs. METHODS Using an optimized algorithm for automated detection, all CASEs were detected in a population of 10 premature infants developing late-onset sepsis and 10 age-matched control patients. CASEs were inspected by two clinical experts and annotated as CAs or rejections in two rounds of annotations. A total of 47 features were extracted from the ECG, CI and oxygen saturation signals considering four 30 s-long moving windows, from 30 s before to 15 s after the onset of each CASE, using a moving step size of 5 s. Consecutively, new CA detection models were developed based on logistic regression with elastic net penalty, random forest and support vector machines. Performance was evaluated using both leave-one-patient-out and 10-fold cross-validation considering the mean area under the receiver-operating-characteristic curve (AUROC). RESULTS The CA detection model based on logistic regression with elastic net penalty returned the highest mean AUROC when features extracted from all four time windows were included, both using leave-one-patient-out and 10-fold cross-validation (mean AUROC of 0.88 and 0.90, respectively). Feature relevance was found to be the highest for features derived from the CI. A threshold for the false positive rate in the mean receiver-operating-characteristic curve equal to 0.3 led to a high percentage of correct detections for all CAs (78.2%) and even higher for CAs followed by a bradycardia (93.4%) and CAs followed by both a bradycardia and a desaturation (95.2%), which are more critical for the well-being of premature infants. CONCLUSIONS Models based on machine learning can lead to improved CA detection with fewer false alarms.
Collapse
Affiliation(s)
- Gabriele Varisco
- Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands; Clinical Physics, Máxima Medical Center, Veldhoven, the Netherlands.
| | - Zheng Peng
- Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands; Clinical Physics, Máxima Medical Center, Veldhoven, the Netherlands
| | - Deedee Kommers
- Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands; Pediatrics, Máxima Medical Center, Veldhoven, the Netherlands
| | - Zhuozhao Zhan
- Mathematics and Computer Science, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Ward Cottaar
- Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Peter Andriessen
- Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands; Pediatrics, Máxima Medical Center, Veldhoven, the Netherlands
| | - Xi Long
- Philips Research, Eindhoven, the Netherlands; Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Carola van Pul
- Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands; Clinical Physics, Máxima Medical Center, Veldhoven, the Netherlands
| |
Collapse
|
37
|
Yang WC, Fogel A, Lauria ME, Ferguson K, Smith ER. Fast Feed Advancement for Preterm and Low Birth Weight Infants: A Systematic Review and Meta-analysis. Pediatrics 2022; 150:188645. [PMID: 35921676 DOI: 10.1542/peds.2022-057092g] [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] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Fast feed advancement may reduce hospital stay and infection but may increase adverse outcomes in preterm and low birth weight infants. The objective of this study was to assess effects of fast feed advancement (≥30 ml/kg per day) compared with slow feed advancement (<30 ml/kg per day) in preterm and low birth weight infants. METHODS Data sources include Medline, Scopus, Web of Science, CINAHL, and Index Medicus through June 30, 2021. Randomized trials were selected. Primary outcomes were mortality, morbidity, growth, and neurodevelopment. Data were extracted and pooled using random-effects models. The Cochrane Risk of Bias 2 tool was used. RESULTS A total of 12 RCTs with 4291 participants were included. At discharge, there was moderate certainty evidence that fast advancement likely slightly reduces the risk of: mortality (relative risk [RR] 0.93, 95% confidence interval [95% CI] 0.73 to 1.18, I2 = 18%, 11 trials, 4132 participants); necrotizing enterocolitis (RR 0.89, 95% CI 0.68 to 1.15, I2 = 0%, 12 trials, 4291 participants); sepsis (RR 0.92, 95% CI 0.83 to 1.03, I2 = 0%, 9 trials, 3648 participants); and feed intolerance (RR 0.92, 95% CI 0.77 to 1.10, I2 = 0%, 8 trials, 1114 participants). Fast feed advancement may also reduce the risk of apnea (RR 0.72, 95% CI 0.47 to 1.12, I2 = 0%, low certainty, 2 trials, 153 participants). Fast feed advancement decreases time to regain birth weight (mean difference [MD] -3.69 days, 95% CI -4.44 to -2.95, I2 = 70%, high certainty, 6 trials, 993 participants,) and likely reduces the duration of hospitalization (MD -3.08 days, 95% CI -4.34 to -1.81, I2 = 77%, moderate certainty, 7 trials, 3864 participants). Limitations include heterogeneity between studies and small sample sizes. CONCLUSIONS Fast feed advancement reduces time to regain birth weight and likely reduces the length of hospital stay; it also likely reduces the risk of neonatal morbidity and mortality slightly. However, it may increase the risk of neurodevelopmental disability slightly. More studies are needed to understand the long-term effects of fast feed advancement.
Collapse
Affiliation(s)
- Wen-Chien Yang
- The George Washington University, Milken Institute School of Public Health, Washington, District of Columbia
| | | | - Molly E Lauria
- The George Washington University, Milken Institute School of Public Health, Washington, District of Columbia.,ICF, Rockville, Maryland
| | - Kacey Ferguson
- The George Washington University, Milken Institute School of Public Health, Washington, District of Columbia
| | - Emily R Smith
- The George Washington University, Milken Institute School of Public Health, Washington, District of Columbia
| |
Collapse
|
38
|
Necrotizing enterocolitis in preterm newborn with a history of maternal COVID-19: a case report. Radiol Case Rep 2022; 17:2630-2634. [PMID: 35663817 PMCID: PMC9160280 DOI: 10.1016/j.radcr.2022.04.056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
A 6-day-old newborn male presented with a primary complaint of abdominal distension for 3 days before hospital admission. The newborn was delivered from a confirmed COVID-19–positive mother through caesarean section at 38-39 weeks with a birth weight of 2800 grams. Various tests, including blood tests, cultures, and radiography were performed. The patient was diagnosed with necrotizing enterocolitis and sepsis; the prevalence of necrotizing enterocolitis in at-term newborns with >36 weeks of gestational age is 10%. In this case, NEC was suggested to be due to maternal infection during the prenatal phase, as the mother was a confirmed COVID-19 patient. Further exploration is needed to illustrate neonatal outcomes following necrotizing enterocolitis diagnosis in correlation with maternal intrauterine COVID-19 infection.
Collapse
|
39
|
Fecal amine metabolite analysis before onset of severe necrotizing enterocolitis in preterm infants: a prospective case-control study. Sci Rep 2022; 12:12310. [PMID: 35853977 PMCID: PMC9296556 DOI: 10.1038/s41598-022-16351-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 07/08/2022] [Indexed: 11/08/2022] Open
Abstract
Infants developing necrotizing enterocolitis (NEC) have a different metabolomic profile compared to controls. The potential of specific metabolomics, i.e. amino acids and amino alcohols (AAA), as early diagnostic biomarkers for NEC is largely unexplored. In this multicenter prospective case-control study, longitudinally collected fecal samples from preterm infants (born <30 weeks of gestation) from 1-3 days before diagnosis of severe NEC (Bell's stage IIIA/IIIB), were analyzed by targeted high-performance liquid chromatography (HPLC). Control samples were collected from gestational and postnatal age-matched infants. Thirty-one NEC cases (15 NEC IIIA;16 NEC IIIB) with 1:1 matched controls were included. Preclinical samples of infants with NEC were characterized by five increased essential amino acids-isoleucine, leucine, methionine, phenylalanine and valine. Lysine and ethanolamine ratios were lower prior to NEC, compared to control samples. A multivariate model was rendered based on isoleucine, lysine, ethanolamine, tryptophan and ornithine, modestly discriminating cases from controls (AUC 0.67; p < 0.001). Targeted HPLC pointed to several specific AAA alterations in samples collected 1-3 days before NEC onset, compared to controls. Whether this reflects metabolic alterations and has a role in early biomarker development for NEC, has yet to be elucidated.
Collapse
|
40
|
Shi Z, Guan N, Sun W, Sun T, Niu L, Li J, Ge J. Protective Effect of Levilactobacillus brevis Against Yersinia enterocolitica Infection in Mouse Model via Regulating MAPK and NF-κB Pathway. Probiotics Antimicrob Proteins 2022; 14:830-844. [PMID: 35665480 DOI: 10.1007/s12602-022-09957-x] [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] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
Although the use of the probiotic bacterium Lactobacillus for the treatment and prevention of diseases caused by various pathogenic bacteria has received increasing attention in recent years, its mechanism remains incompletely understood. Levilactobacillus brevis 23017 is a select probiotic strain that can regulate the immunity of host animals and resist pathogen infections. In this study, we analyzed the effect of L. brevis 23017 on Yersinia enterocolitica intestinal infection in a BALB/c mouse model and discussed its underlying mechanism. We found that in the mouse model, L. brevis 23017 prevented the damage of villi in the small intestine and decelerated weight loss after Y. enterocolitica infection. Moreover, we focused on the mechanism of the protective effect of L. brevis 23017 from the perspective of the damage and repair of the intestinal mucosal barrier. We observed that L. brevis 23017 maintained a normal mucosal barrier by altering the expression of tight junction proteins. Notably, our results indicated that L. brevis 23017 effectively promoted the secretion of the intestine-specific secretory immunoglobulin A (SIgA) by B cells via regulating cytokines and oxidative damage levels. This mechanism may be the reason for its protective role in Y. enterocolitica infection. In addition, our results demonstrated that the mechanism of L. brevis 23017 was related to antibacterial colonization and inflammation regulation and closely related to antioxidative stress and SIgA promotion. The protective effect of L. brevis 23017 on mice was related to the signaling pathway protein p38 MAPK and the phosphorylation levels of NF-κB. Our study provided novel insight into the mechanism of Lactobacillus against pathogenic bacterial infections. Such insight is of great importance for the prevention, diagnosis, and treatment of related diseases.
Collapse
Affiliation(s)
- Ziqi Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Naiyu Guan
- Key Laboratory of Zoonoses Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Weijiao Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tianzhi Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Lingdi Niu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jinyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
- Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin, 150030, China.
| |
Collapse
|
41
|
Cai X, Golubkova A, Hunter CJ. Advances in our understanding of the molecular pathogenesis of necrotizing enterocolitis. BMC Pediatr 2022; 22:225. [PMID: 35468817 PMCID: PMC9036771 DOI: 10.1186/s12887-022-03277-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/08/2022] [Indexed: 11/24/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a multifactorial and complex disease. Our knowledge of the cellular and genetic basis of NEC have expanded considerably as new molecular mechanisms have been identified. This article will focus on the current understanding of the molecular pathogenesis of NEC with a focus on the inflammatory, immune, infectious, and genetic mechanisms that drive disease development.
Collapse
Affiliation(s)
- Xue Cai
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Alena Golubkova
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Catherine J Hunter
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
42
|
Nesterenko TH, Baliga N, Swaintek S, Abdelatif D, Aly H, Mohamed MA. The impact of a multifaceted quality improvement program on the incidence of necrotizing enterocolitis in very low birth weight infants. Pediatr Neonatol 2022; 63:181-187. [PMID: 34933821 DOI: 10.1016/j.pedneo.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/27/2021] [Accepted: 10/07/2021] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a multifactorial gastrointestinal disease which mostly occurs in very low birth weight (VLBW) infants. In addition to decreasing gestational age (GA) or birth weight (BW), artificial formula, delayed initiation or rapidly advanced feeding, severe anemia and systemic infections were associated with NEC. Several studies demonstrated that breast milk, standardized feeding advancement regimens and treatment of anemia are associated with less incidence of NEC. It is not known if including all these interventions in one multifaceted program will lead to significant reduction in NEC. METHODS The NICU team at The George Washington University Hospital created a multifaceted interdisciplinary quality improvement project to tackle several aspects of NEC prevention that addressed researched risk factors for NEC. The program was made of four quality improvement protocols: 1) Standardized Structured Feeding Program, 2) Feeding Intolerance Management Algorithm, 3) Enteral Osmolality Control Tool, and 4) Packed Red Blood Cell (RBC) Standardized Transfusion Protocol. This time-series, quasi experimental study design examined the differences in the incidence of NEC between infants with BW < 1500 g who were admitted to the GW Hospital NICU before and after the program implementation. RESULTS Data from 408 VLBW infants were included in the study. Although not statistically significant, there was a decreasing trend of NEC incidence in the post-implementation group (n = 199) compared to the pre-implementation group (n = 209), (3.5% vs. 5.3%, p = 0.88). The trend in the incidence of NEC declined further after the introduction of RBC transfusion protocol which was introduced ten month after starting the other elements of the program. CONCLUSION Integration of the multifaceted quality improvement program may be associated with a decline in the occurrence of NEC. Further analysis with a larger sample size is required to determine if the changes seen are statistically significant.
Collapse
Affiliation(s)
- Tetyana H Nesterenko
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, OH, USA
| | - Nita Baliga
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Sarah Swaintek
- Department of Food and Nutrition, The George Washington University Hospital, Washington, DC, USA
| | - Dinan Abdelatif
- Department of Obstetrics and Gynecology, The George Washington University, Washington, DC, USA
| | - Hany Aly
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, OH, USA
| | - Mohamed A Mohamed
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, OH, USA.
| |
Collapse
|
43
|
Lueschow SR, Boly TJ, Frese SA, Casaburi G, Mitchell RD, Henrick BM, McElroy SJ. Bifidobacterium longum Subspecies infantis Strain EVC001 Decreases Neonatal Murine Necrotizing Enterocolitis. Nutrients 2022; 14:495. [PMID: 35276854 PMCID: PMC8839161 DOI: 10.3390/nu14030495] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 01/04/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a disease mainly of preterm infants with a 30-50% mortality rate and long-term morbidities for survivors. Treatment strategies are limited and have not improved in decades, prompting research into prevention strategies, particularly with probiotics. Recent work with the probiotic B. infantis EVC001 suggests that this organism may generate a more appropriate microbiome for preterm infants who generally have inappropriate gut colonization and inflammation, both risk factors for NEC. Experimental NEC involving Paneth cell disruption in combination with bacterial dysbiosis or formula feeding was induced in P14-16 C57Bl/6 mice with or without gavaged B. infantis. Following completion of the model, serum, small intestinal tissue, the cecum, and colon were harvested to examine inflammatory cytokines, injury, and the microbiome, respectively. EVC001 treatment significantly decreased NEC in a bacterial dysbiosis dependent model, but this decrease was model-dependent. In the NEC model dependent on formula feeding, no difference in injury was observed, but trending to significant differences was observed in serum cytokines. EVC001 also improved wound closure at six and twelve hours compared to the sham control in intestinal epithelial monolayers. These findings suggest that B. infantis EVC001 can prevent experimental NEC through anti-inflammatory and epithelial barrier restoration properties.
Collapse
Affiliation(s)
- Shiloh R. Lueschow
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Timothy J. Boly
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven A. Frese
- Department of Nutrition, University of Nevada, Reno, NV 89557, USA;
| | - Giorgio Casaburi
- Department of Bioinformatics, Metabiomics, Carlsbad, CA 92008, USA;
| | - Ryan D. Mitchell
- Evolve Biosystems, Inc., Davis, CA 95618, USA; (R.D.M.); (B.M.H.)
| | - Bethany M. Henrick
- Evolve Biosystems, Inc., Davis, CA 95618, USA; (R.D.M.); (B.M.H.)
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
| | - Steven J. McElroy
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
44
|
Chandramowlishwaran P, Raja S, Maheshwari A, Srinivasan S. Enteric Nervous System in Neonatal Necrotizing Enterocolitis. Curr Pediatr Rev 2022; 18:9-24. [PMID: 34503418 DOI: 10.2174/1573396317666210908162745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/26/2021] [Accepted: 06/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The pathophysiology of necrotizing enterocolitis (NEC) is not clear, but increasing information suggests that the risk and severity of NEC may be influenced by abnormalities in the enteric nervous system (ENS). OBJECTIVE The purpose of this review was to scope and examine the research related to ENS-associated abnormalities that have either been identified in NEC or have been noted in other inflammatory bowel disorders (IBDs) with histopathological abnormalities similar to NEC. The aim was to summarize the research findings, identify research gaps in existing literature, and disseminate them to key knowledge end-users to collaborate and address the same in future studies. METHODS Articles that met the objectives of the study were identified through an extensive literature search in the databases PubMed, EMBASE, and Scopus. RESULTS The sources identified through the literature search revealed that: (1) ENS may be involved in NEC development and post-NEC complications, (2) NEC development is associated with changes in the ENS, and (3) NEC-associated changes could be modulated by the ENS. CONCLUSION The findings from this review identify the enteric nervous as a target in the development and progression of NEC. Thus, factors that can protect the ENS can potentially prevent and treat NEC and post-NEC complications. This review serves to summarize the existing literature and highlights a need for further research on the involvement of ENS in NEC.
Collapse
Affiliation(s)
- Pavithra Chandramowlishwaran
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - Shreya Raja
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Shanthi Srinivasan
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
45
|
Angelika D, Etika R, Fitriah M, Kusumawardani NN, Vita AD, Irawan R, Liem KD, Ugrasena IDG. Association between glial fibrillary acidic protein, glial-derived neurotrophic factor, and fatty acid-binding protein-2 at birth in the incidence of necrotizing enterocolitis in preterm infants. Front Pediatr 2022; 10:1010013. [PMID: 36340713 PMCID: PMC9630751 DOI: 10.3389/fped.2022.1010013] [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: 08/02/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND This study aimed to analyze the relationship between glial fibrillary acidic protein (GFAP), glial-derived neurotrophic factor (GDNF), and fatty acid-binding protein-2 (FABP-2) in preterm infants on the incidence of NEC. METHODS Preterm infants with a birth weight <1,500 g and gestational age <34 weeks were included in this study. Biomarker examination was performed using the umbilical vein blood at birth (first sample). Biomarker examination was repeated if the infant developed symptoms of NEC using peripheral vein blood (second sample). Infants were observed for 14 days. If NEC did not exist, a biomarker examination was performed at 14 days. RESULTS This study included 30 preterm infants, nine infants experienced NEC. The values of GFAP, GDNF, and FABP-2 (median and range) in the group with NEC were higher than those in the group without NEC in both the first samples {GFAP [1.40 (0.20-6.50) vs. 0.30 (0.10-1.30) P = 0.014], GDNF [2.84 (1.05-14.11) vs. 1.56 (1.07-3.48) P = 0.050], and FABP-2 [621.70 (278.40-2,207.00) vs. 294.20 (211.40-597.50) P = 0.002]} and second samples {GFAP [2.40 (0.30-3.10) vs. 0.30 (0.10-0.60) P = 0.003], GDNF [2.99 (0.56-10.30) vs. 1.46 (0.85-2.24) P = 0.019], and FABP-2 [646.8 (179.20-1,571.00) vs. 314.90 (184.70-521.60) P = 0.040]}. In infants with NEC, the median values of GFAP [2.40 (0.30-3.10) vs. 1.40 (0.20-6.50) P = 0.767], GDNF [2.99 (0.56-10.30) vs. 2.84 (1.05-14.11) P = 0.859], and FABP-2 [646.80 (179.20-1,571.00) vs. 621.70 (278.40-2,207.00) P = 0.953] in the second sample were higher than those in the first sample. Logistic regression demonstrated that GFAP at birth (Odds Ratio [OR] = 15.629, 95% Confidence Interval [CI] = 1.697-143.906, P = 0.015) and FABP-2 levels at birth (OR = 1.008, 95% CI = 1.001-1.015, P = 0.033) were significantly associated with an increased risk of NEC. CONCLUSION Increased GFAP, GDNF, and FABP-2 at birth are associated with NEC occurrence within two weeks of birth. These findings suggest that early-onset NEC is associated with intestinal injury that occurs during the perinatal or even prenatal period.
Collapse
Affiliation(s)
- Dina Angelika
- Doctoral Program of Medical Science, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Risa Etika
- Department of Child Health, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Munawaroh Fitriah
- Department of Clinical Pathology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | | | - Angelica Diana Vita
- Medical Program, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Roedi Irawan
- Department of Child Health, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Kian Djien Liem
- Department of Neonatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Dewa Gede Ugrasena
- Department of Child Health, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
46
|
An Interdisciplinary Approach to Reducing NEC While Optimizing Growth: A 20-Year Journey. Adv Neonatal Care 2021; 21:433-442. [PMID: 34510070 DOI: 10.1097/anc.0000000000000929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) and postnatal growth restriction are significant clinical dilemmas that contribute to short- and long-term morbidities for the most premature infants. PURPOSE After a rise in NEC rates in a regional neonatal intensive care unit (NICU), improvement practices were implemented by an interdisciplinary quality improvement (QI) work group whose focus was initially on nutrition and growth. QI work was refocused to address both NEC and growth concurrently. METHODS Through various QI initiatives and with evolving understanding of NEC and nutrition, the work group identified and implemented multiple practices changes over 2-decade time span. A standardized tool was used to review each case of NEC and outcomes were continually tracked to guide QI initiatives. LOCAL FINDINGS Focused QI work contributed to a significant reduction in NEC rates from 16.2% in 2007 to 0% in 2018 for inborn infants. Exclusive human milk diet was a critical part of the success. Postnatal growth outcomes initially declined after initial NEC improvement work. Improvement work that focused jointly on NEC and nutrition resulted in improved growth outcomes without impacting NEC. IMPLICATIONS FOR PRACTICE Use of historical perspective along with evolving scientific understanding can guide local improvement initiatives. Work must continue to optimize lactation during NICU hospitalization. More research is needed to determine impact of care practices on gastrointestinal inflammation including medication osmolality, probiotics, and noninvasive respiratory support.
Collapse
|
47
|
Vagal Tone and Proinflammatory Cytokines Predict Feeding Intolerance and Necrotizing Enterocolitis Risk. Adv Neonatal Care 2021; 21:452-461. [PMID: 34847103 DOI: 10.1097/anc.0000000000000959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is the leading cause of death due to gastrointestinal disease in preterm neonates; yet, clinicians lack reliable and noninvasive predictive tools. PURPOSE We aimed to test that diminished high-frequency heart rate variability (HF-HRV) and elevated levels of proinflammatory cytokines would have utility in NEC prediction. METHODS In this multisite prospective study, we enrolled 250 preterm (26-34 weeks' postmenstrual age [PMA]) neonates with physiological stability at 72 hours of life. HRV was measured noninvasively using electrocardiograhic data from standardized cardiorespiratory monitors at postnatal week 1 of life and weekly thereafter until 35 weeks' PMA or discharge; blood was collected for cytokines at postnatal weeks 1 and 3. NEC was diagnosed via Modified Bell's Staging Criteria. RESULTS HF-HRV was decreased at weeks 1 and 2 in neonates (47% females) who developed feeding intolerance or stage 2+ NEC. In addition, these neonates displayed elevated levels of IL-8 at week 1 and increased levels of IL-1β, IL-6, TNF-α, and IL-8 at week 3 of life. Low HF-HRV was associated with elevated IL-6 or IL-8 levels at weeks 1 and 3 of life. Logistic regression indicated that only HF-HRV was a significant predictor of feeding intolerance or NEC development. IMPLICATIONS FOR PRACTICE AND RESEARCH HRV is a promising noninvasive modality for NEC risk detection. The association of low HF-HRV with elevated proinflammatory cytokines provides evidence for a putative role of the vagal cholinergic pathway in NEC pathogenesis. Future studies should focus on application of these techniques to test clinical therapeutics.Video Abstract available at https://journals.lww.com/advancesinneonatalcare/Pages/videogallery.aspx?autoPlay=false&videoId=54.
Collapse
|
48
|
Blood group AB is associated with poor outcomes in infants with necrotizing enterocolitis. J Pediatr Surg 2021; 56:1911-1915. [PMID: 34392969 DOI: 10.1016/j.jpedsurg.2021.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/25/2021] [Accepted: 07/13/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a neonatal disease associated with necrosis and perforation of the bowel. We investigated the association between blood group and NEC outcomes and the potential contribution of fetal-maternal blood group incompatibility. METHODS Retrospective study including all preterm-born infants with NEC (≥ Bell's stage IIa) admitted to our NICU between January 2008 and October 2019. We analyzed the association between infants' blood groups and fetal-maternal blood group incompatibility with Bell stage severity, need for surgery, and mortality due to NEC. RESULTS We included 237 NEC patients. In univariable analyses both AB blood group and fetal-maternal blood group incompatibility increased infants' risk of severe outcomes, with odds ratios (OR) ranging from 6.57 to 12.06 and 1.97 to 2.38, respectively. When adjusted for gestational age only AB blood group remained significant with OR 7.47 (95% confidence interval, 1.95-28.53, P = 0.003), 12.37 (2.63-58.20, P = 0.001), and 8.16 (2.28-29.14, P = 0.001) for NEC Bell's stage III, need for surgery, and NEC related mortality, respectively. Blood group incompatibility adjusted for gestational age was not related to worse outcomes with OR 1.84 (0.87-3.89, P = 0.11, 2.08 (0.98-4.41, P = 0.06) 1.52 (0.68-3.42, P = 0.31), for NEC Bell's stage III, need for surgery, and NEC related mortality, respectively. CONCLUSION Our data confirm an association between blood group AB and worse outcomes in NEC infants, but this is not based on fetal-maternal blood group incompatibility.
Collapse
|
49
|
Al-Alaiyan S, Abdulaziz N, Alkohlani A, Almairi SO, Al Hazzani F, Binmanee A, Alfattani A. Effects of Probiotics and Lactoferrin on Necrotizing Enterocolitis in Preterm Infants. Cureus 2021; 13:e18256. [PMID: 34712533 PMCID: PMC8542402 DOI: 10.7759/cureus.18256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 12/29/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in neonates. Despite intensive research, the etiology and pathophysiology of NEC is still obscure. Evidence from recent studies and meta-analyses showed a significant role of probiotics as a prophylactic measure in reducing NEC, sepsis, and mortality. However, obstacles against the generalization of the results still remain. The aim of the study was to evaluate the role of prophylactic administration of probiotics and lactoferrin in reducing the rate of NEC in preterm infants. Methods In this retrospective cohort study, all medical records of infants born with a birth weight of 1,500 g and less who were born between 2012 and 2017 were reviewed. The enrolled infants were divided into two groups: group 1 included infants born between January 2012 and August 2014, a period before probiotics were started in our unit, and group 2 included infants born between January 2014 and December 2017 after starting probiotics and lactoferrin. Multiple variables were collected including maternal data, neonatal data, and risk factors for NEC. Results Medical records of 284 infants who met our inclusion criteria were reviewed. Of the 284 infants, 134 were in group 1 and 150 infants were in group 2. There were no significant statistical differences between group 1 and group 2 in neonatal and maternal demographic data and clinical data. Of 134 infants who received probiotics and lactoferrin, 11 developed NEC, while 26 of the 150 infants in group 2 developed NEC, and the difference was statistically significant (p = 0.023). Conclusion Probiotics and lactoferrin given orally to very low birth weight preterm infants were associated with a decreased rate of NEC.
Collapse
Affiliation(s)
- Saleh Al-Alaiyan
- Pediatrics/Neonatal-Perinatal Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, SAU.,Pediatrics, Alfaisal University, Riyadh, SAU
| | - Najlaa Abdulaziz
- Pediatrics, King Faisal Specialist Hospital & Research Centre, Riyadh, SAU
| | | | - Sana O Almairi
- Pediatrics, Alfaisal University College of Medicine, Riyadh, SAU
| | - Fahad Al Hazzani
- Pediatrics, King Faisal Specialist Hospital & Research Centre, Riyadh, SAU
| | - Abdulaziz Binmanee
- Pediatrics, King Faisal Specialist Hospital & Research Centre, Riyadh, SAU
| | - Areej Alfattani
- Biostatistics and Epidemiology, King Faisal Specialist Hospital & Research Centre, Riyadh, SAU
| |
Collapse
|
50
|
Jeziorczak PM, Frenette RS, Aprahamian CJ. Lack of Enteral Feeding Associated with Mortality in Prematurity and Necrotizing Enterocolitis. J Surg Res 2021; 270:266-270. [PMID: 34715538 DOI: 10.1016/j.jss.2021.09.028] [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: 03/03/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Necrotizing Enterocolitis (NEC) remains a significant cause of morbidity and mortality. Recently, there has been an increased recognition of the importance of intestinal immunity and the associations with antibiotics and enteral feeds in the pathophysiology of NEC. The primary purpose of this study is to examine the association of enteral feeds on the survival of premature neonates with NEC. MATERIAL AND METHODS A retrospective review using the Vermont Oxford Network for a Level IV NICU from January 1, 2013 through December 31, 2019 was performed. All neonates had a gestational age between 22 to 29 weeks, weighed at least 300 grams (n = 653), had a reported enteral feed status and were treated for NEC (n = 43). Data analysis utilized two-tailed t-tests for NEC and infection rates then Fisher's exact tests for survival status. RESULTS The incidence of NEC in the population was 6.6% (43/653). Of the 43 neonates treated for NEC, 27 were enterally fed, while the other 16 were not. All 27 neonates with NEC that were able to achieve enteral feeds survived and had an infection rate of 22.2%. Meanwhile, all 16 neonates with NEC that were unable to achieve enteral feeds died and had an infection rate of 62.5%. CONCLUSIONS There is a significant association between enteral feeds and NEC, survival, and infection rates in premature neonates. These findings support the importance of intestinal immunity and the microbiota in NEC. Given the limitations of the retrospective review, the profound survival advantage with enteral feeds reinforces the need for further study.
Collapse
Affiliation(s)
- Paul M Jeziorczak
- OSF Healthcare- Children's Hospital of Illinois, Peoria, IL; University of Illinois College of Medicine at Peoria, Peoria, IL
| | - Riley S Frenette
- OSF Healthcare- Children's Hospital of Illinois, Peoria, IL; A.T. Still University-Kirksville College of Osteopathic Medicine, Kirksville, MO.
| | - Charles J Aprahamian
- OSF Healthcare- Children's Hospital of Illinois, Peoria, IL; University of Illinois College of Medicine at Peoria, Peoria, IL
| |
Collapse
|