1
|
El Manouni El Hassani S, Frerichs NM, Berkhout DJC, Nijsen T, Knobel HH, Weda H, Xu M, Long X, Wijnoltz L, van Weissenbruch MM, van Kaam AH, Cossey V, Peeters CFW, van Lingen RA, Hulzebos CV, Vijlbrief DC, de Boode WP, Kramer BW, Budding AE, Benninga MA, de Boer NKH, Niemarkt HJ, de Meij TGJ. Longitudinal fecal microbiota and volatile metabolomics preceding necrotizing enterocolitis in preterm infants: a case-control study. Sci Rep 2025; 15:10419. [PMID: 40140438 PMCID: PMC11947453 DOI: 10.1038/s41598-025-94692-w] [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: 08/08/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Alterations in fecal microbiota and volatile organic compound (VOC) profiles of preterm infants have been demonstrated before onset of necrotizing enterocolitis (NEC). However, NEC-specific signatures need to be identified before potential application as predictive biomarker in clinical practice. A prospective multicenter case-control study was conducted to identify preclinical fecal microbiota and VOC profiles of infants that developed NEC. Microbiota analysis (PCR-based IS-pro technique) and VOC analysis (gas chromatography-mass spectrometry) were performed on fecal samples collected up to three days before clinical NEC onset. In 112 infants (56 NEC, 56 matched controls), sufficient number fecal samples were collected for either microbiota or VOC analysis. Prior to NEC onset, Clostridium perfringens (p = 0.023, unadjusted) was more present in infants with NEC, versus controls. VOC analysis showed a clear distinction between fecal profiles of NEC cases and controls (area under the curve = 0.82). Fourteen unique VOC features contributed to this discrimination. Fecal microbiota and VOC profiles may serve as early indicators of NEC, and allow for increased understanding of pathophysiological mechanisms of NEC, but larger validation cohorts are needed before an overarching NEC-specific predictive microbiota-based biomarker can be implemented.
Collapse
Affiliation(s)
- S El Manouni El Hassani
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - N M Frerichs
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, The Netherlands.
| | - D J C Berkhout
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - T Nijsen
- Philips S&RC Research & Development, Eindhoven, The Netherlands
| | - H H Knobel
- Eurofins EAG, Eurofins Materials Science Netherlands B.V., Eindhoven, The Netherlands
| | - H Weda
- Netherlands Institute for Public Safety, Arnhem, The Netherlands
| | - M Xu
- Lighting and IoT Lab, Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - X Long
- Biomedical Diagnostics Lab, Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - L Wijnoltz
- Philips Research, Eindhoven, The Netherlands
| | - M M van Weissenbruch
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neonatology, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - A H van Kaam
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neonatology, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - V Cossey
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - C F W Peeters
- Mathematical and Statistical Methods Group (Biometris), Wageningen University & Research, Wageningen, The Netherlands
| | - R A van Lingen
- Neonatal Intensive Care Unit, Amalia Children's Centre, Isala, Zwolle, The Netherlands
| | - C V Hulzebos
- Neonatal Intensive Care Unit, Beatrix Children's Hospital/University Medical Centre Groningen, Groningen, The Netherlands
| | - D C Vijlbrief
- Neonatal Intensive Care Unit, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - W P de Boode
- Neonatal Intensive Care Unit, Radboud University Medical Centre, Radboud Institute for Health Sciences, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - B W Kramer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - M A Benninga
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - N K H de Boer
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - H J Niemarkt
- Neonatal Intensive Care Unit, Máxima Medical Center, Veldhoven, The Netherlands
| | - T G J de Meij
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Cai J, Yan X, Liu X, Yin X, Shi A, Ji C, Cao Y. Human β-casein-derived peptide BCCY-1 improved the intestinal barrier integrity by regulating the TLR4/eNOS/3-Nitrotyrosine axis. Food Chem 2025; 463:140821. [PMID: 39244994 DOI: 10.1016/j.foodchem.2024.140821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024]
Abstract
Necrotizing enterocolitis (NEC) is a lethal gastrointestinal disease affecting premature infants. Although earlier studies have highlighted protective effects of milk-derived peptides against NEC, the role of the human β-casein-derived peptide BCCY-1 in intestinal barrier protection has never been investigated. Here, we showed that BCCY-1 alleviated the phenotype of NEC, reduced intestinal expression of Toll-like receptor 4 (TLR4) and interleukin-6, and improved the intestinal barrier integrity. NEC-associated multi-organ injury and impaired bone marrow hematopoiesis were also attenuated by BCCY-1. Metabolic screening revealed significant changes in intestinal metabolites in the NEC and NEC + BCCY-1 groups. Further analysis disclosed inhibition of 3-Nitrotyrosine formation due to the preservation of endothelial nitric oxide synthase (eNOS) activity, which was associated with the interactions between BCCY-1 and lipopolysaccharides, leading to disruption of TLR4 signaling. Our findings suggested that BCCY-1 improved intestinal barrier integrity through modulating the TLR4/eNOS/3-Nitrotyrosine axis, highlighting its potential role in the maintenance of intestinal health.
Collapse
Affiliation(s)
- Jinyang Cai
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xiangyun Yan
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China
| | - Xinyue Liu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China
| | - Xiaoxiao Yin
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China
| | - Aiwu Shi
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China
| | - Chenbo Ji
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China.
| | - Yan Cao
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, Jiangsu, China.
| |
Collapse
|
3
|
Wolski M, Ciesielski T, Buczma K, Fus Ł, Girstun A, Trzcińska-Danielewicz J, Cudnoch-Jędrzejewska A. Administration of Adipose-Derived Stem Cells After the Onset of the Disease Does Not Lower the Levels of Inflammatory Cytokines IL1 and IL6 in a Rat Model of Necrotizing Enterocolitis. Biomedicines 2024; 12:2897. [PMID: 39767803 PMCID: PMC11727438 DOI: 10.3390/biomedicines12122897] [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: 11/12/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Research on the roles of stem cells in necrotizing enterocolitis (NEC) has primarily focused on the effects of bone marrow- and amniotic fluid-derived stem cells in mitigating the clinical manifestations of the disease. However, the potential of adipose tissue-derived stem cells (ADSCs) remains unexplored in this context. The aim of this study was to evaluate the therapeutic potential of ADSC administration during the active inflammatory phase of NEC, with a specific focus on reducing the levels of the inflammatory cytokines IL-1 and IL-6. Methods: A self-modified hypoxia-hypothermia-formula feeding rat NEC model was employed. A total of 117 rat pups were divided into two groups: a treatment group (NEC-ADSC, n = 55) and a control group (NEC-PLCB (placebo), n = 62). In the NEC-ADSC group, ADSCs were administered intraperitoneally 24 h into the NEC protocol. After 72 h, bowel and fluid samples were collected for analysis. Results: The analysis revealed no significant effect on NEC histopathology (p = 0.347) or on the levels of IL-1 and IL-6 (p = 0.119 and p = 0.414, respectively). Conclusions: The administration of adipose tissue-derived stem cells after the onset of necrotizing enterocolitis does not reduce the levels of inflammatory cytokines IL-1 and IL-6, nor does it influence the histopathological outcomes of the disease in the rat model. Further research is needed to explore the potential therapeutic role of adipose tissue-derived stem cells in the treatment of necrotizing enterocolitis.
Collapse
Affiliation(s)
- Marek Wolski
- Department of Pediatric Surgery, Medical University of Warsaw, Zwirki i Wigury 63a, 02-091 Warsaw, Poland
| | - Tomasz Ciesielski
- Laboratory of Centre for Preclinical Research, Chair and Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (T.C.); (K.B.); (A.C.-J.)
| | - Kasper Buczma
- Laboratory of Centre for Preclinical Research, Chair and Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (T.C.); (K.B.); (A.C.-J.)
| | - Łukasz Fus
- Department of Pathology, Medical University of Warsaw, Pawinskiego 7, 02-106 Warsaw, Poland;
| | - Agnieszka Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (A.G.); (J.T.-D.)
| | - Joanna Trzcińska-Danielewicz
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (A.G.); (J.T.-D.)
| | - Agnieszka Cudnoch-Jędrzejewska
- Laboratory of Centre for Preclinical Research, Chair and Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (T.C.); (K.B.); (A.C.-J.)
| |
Collapse
|
4
|
Wei J, Tian Y, Guan M, Wei J, Ji Y, Tao G, Sylvester KG. Sodium formate induces development-dependent intestinal epithelial injury via necroptosis and apoptosis. Redox Rep 2024; 29:2433393. [PMID: 39620924 PMCID: PMC11613409 DOI: 10.1080/13510002.2024.2433393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES Necrotizing enterocolitis (NEC) is a common and sometimes fatal disease affecting premature infants. Elevated formate has been found in the stool of patients with NEC. Sodium formate (NaF) is used to explore the role of formate in the intestinal epithelial injury. METHODS In this study, 150 mM NaF solution was intraluminally injected in 14-day-old and 28-day-old mice. Mice were sacrificed after 24 h of feces collection, and the blood and small intestinal tissues were collected to detect the pathological damage of intestinal tissue, intestinal permeability, oxidative stress indicators including SOD, HO-1, MDA, and 4-HNE, inflammatory cytokines including IL-1β, TNF-α and IL-6, mitochondrial function such as ATP and PGC-1α in mice intestinal tissue, indicators of the cell death modes including necroptosis-related protein RIPK1 and p-MLKL, and apoptosis- related protein cleaved-caspase-3 and p-AKT (S473). RESULTS NaF treatment significantly damaged intestinal epithelial tissue and barrier function, caused mitochondrial dysfunction, manifesting as decreased ATP and PGC-1α levels, increased lipid peroxidation products MDA and 4-HNE, depleted antioxidant enzyme SOD, and upregulated the expression of HO-1. Furthermore, NaF treatment induced inflammatory responses by promoting the release of IL-1β, IL-6 and TNF-α in a development-dependent manner, eventually inducing necroptosis and apoptosis. CONCLUSIONS Formate may be a source of metabolic intestinal injury contributing to the pathogenesis of NEC in human newborns.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yuan Tian
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Meiqi Guan
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jinshu Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yong Ji
- Department of Neonatal Intensive Care Unit, Shanxi Children’s Hospital, Taiyuan, People’s Republic of China
| | - Guozhong Tao
- Pediatric Surgery-Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl G. Sylvester
- Pediatric Surgery-Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
5
|
Zou P, Fang W, Wu L, He J, Xia H, Zhong W, He Q. Identification of risk factors for necrotizing enterocolitis in twins: a case-control matching analysis of over ten-years' experience. BMC Pediatr 2024; 24:744. [PMID: 39548407 PMCID: PMC11568530 DOI: 10.1186/s12887-024-05188-x] [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: 02/20/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVE The identification of risk factors is crucial for the clinical prevention and diagnosis of necrotizing enterocolitis (NEC). Monochorionic twins (MCT), due to the high genetic homogeneity, provided a valuable model for investigating the risk factors of various diseases. This study aimed to explore the risk factors for NEC using MCT. METHODS A retrospective review was conducted on the medical records of monochorionic twins (MCT) treated at Guangzhou Women and Children's Medical Center from January 2012 to March 2023. We compared perinatal condition, feeding and preceding condition between MCT pairs with NEC (NEC MCT) and without NEC(No NEC MCT).Logistic regression analysis was utilized to identify independent risk factors. RESULT In 85 pairs of monochorionic twins (MCT), NEC occurred in one twin in 78.8% of cases, whereas both twins were affected in 21.2% of cases. In the final cohort of 60 pairs of MCT, several parameters were found to differ significantly between NEC MCT group and No NEC MCT group. Compared to No NEC MCT group, the incidence of umbilical cord abnormalities was significantly higher in the NEC MCT group (25% vs. 8.3%, P = 0.014). Meanwhile, NEC MCT group showed higher prevalence of SGA infants (48.3% vs. 21.7%, P = 0.002) and sFGR (38.3% vs. 6.7%, P = 0.000). Furthermore, TTTs (13.3% vs. 3.3%, P = 0.027) and septicemia (25% vs. 5%, P = 0.002) were more common in NEC MCT group. In a multivariable logistic regression model, sFGR (OR 6.81,95%CI 2.1-21.9, p = 0.001) was eventually output as an independent risk factor. CONCLUSION Non-genetic factors play a predominant role in the pathogenesis of NEC. Umbilical cord abnormalities, SGA, sFGR, TTTs and septicemia significantly increase the risk of NEC. sFGR is an independent risk factor of NEC.
Collapse
Affiliation(s)
- Pengjian Zou
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wenhai Fang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Pediatric Surgery, The Maternal and Children Health Care Hospital (Huzhong Hospital) of Huadu, Guangzhou, China
| | - Lili Wu
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Juan He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhong
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiuming He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
6
|
De la Rosa González A, Guerra-Ojeda S, Camacho-Villa MA, Valls A, Alegre E, Quintero-Bernal R, Martorell P, Chenoll E, Serna-García M, Mauricio MD, Serna E. Effect of Probiotics on Gastrointestinal Health Through the Aryl Hydrocarbon Receptor Pathway: A Systematic Review. Foods 2024; 13:3479. [PMID: 39517263 PMCID: PMC11545787 DOI: 10.3390/foods13213479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Probiotics are living microorganisms recognized for conferring health benefits on the host by modulating the gut microbiota. They interact with various signaling pathways, including the aryl hydrocarbon receptor (AhR), which plays a crucial role in maintaining intestinal homeostasis and immune function. The activation of AhR by probiotics has been associated with benefits such as improved intestinal barrier function, reduced inflammation, and modulation of immune responses. This systematic review aims to summarize current knowledge on the signaling of AhR, mediated by probiotics in physiological conditions and gastrointestinal pathologies. We conducted a comprehensive search across databases, including PubMed and Embase, up until July 2024. Out of 163 studies screened, 18 met the inclusion criteria. Our findings revealed in healthy populations that probiotic consumption increases the production of AhR ligands promoting intestinal immune tolerance. Furthermore, in populations with gastrointestinal pathologies, probiotics ameliorated symptoms through AhR activation by Trp metabolites, leading to the upregulation of the anti-inflammatory response.
Collapse
Affiliation(s)
| | - Sol Guerra-Ojeda
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - María Alejandra Camacho-Villa
- HARPEER Research Group, Yumbo 760001, Colombia; (A.D.l.R.G.); (M.A.C.-V.); (R.Q.-B.)
- Pain Study Group (GED), Physical Therapy School, Universidad Industrial de Santander, Bucaramanga Santander 680002, Colombia
| | - Alicia Valls
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- MODULAhR Group, Universitat de Valencia, 46010 Valencia, Spain
| | - Eva Alegre
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
| | | | - Patricia Martorell
- Archer Daniels Midland (ADM), Nutrition, Health & Wellness, Biopolis S. L. Parc Cientific, University of Valencia, 46980 Paterna, Spain; (P.M.); (E.C.)
| | - Empar Chenoll
- Archer Daniels Midland (ADM), Nutrition, Health & Wellness, Biopolis S. L. Parc Cientific, University of Valencia, 46980 Paterna, Spain; (P.M.); (E.C.)
| | - Marta Serna-García
- Department of Dentistry, Faculty of Health Sciences, Universidad Europea de Valencia, 46010 Valencia, Spain;
| | - Maria D. Mauricio
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- MODULAhR Group, Universitat de Valencia, 46010 Valencia, Spain
| | - Eva Serna
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- MODULAhR Group, Universitat de Valencia, 46010 Valencia, Spain
| |
Collapse
|
7
|
Wolski M, Ciesielski T, Buczma K, Fus Ł, Girstun A, Trzcińska-Danielewicz J, Cudnoch-Jędrzejewska A. Administration of Adipose Tissue Derived Stem Cells before the Onset of the Disease Lowers the Levels of Inflammatory Cytokines IL-1 and IL-6 in the Rat Model of Necrotizing Enterocolitis. Int J Mol Sci 2024; 25:11052. [PMID: 39456833 PMCID: PMC11507542 DOI: 10.3390/ijms252011052] [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: 09/15/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
There is little research concerning the role of stem cells in necrotizing enterocolitis (NEC). Bone marrow-derived mesenchymal stem cells (BMDSC) and amniotic fluid-derived stem cells significantly reduced the amount and severity of NEC in the animal models. ADSCs share similar surface markers and differentiation potential with BMDSCs. Their potential role in the setting of NEC has not been researched before. The hypothesis of the study was that prophylactic intraperitoneal administration of ADSCs before the onset of the disease will result in limiting the inflammatory response, effecting a lower incidence of NEC. On a molecular level, this should result in lowering the levels of inflammatory cytokines IL-1 and IL-6. The local ethical committee for animal experiments approval was acquired (WAW2/093/2021). We utilized a self-modified rat NEC model based on single exposure to hypothermia, hypoxia, and formula feeding. One hundred and twenty-eight rat puppies were divided into two groups-prophylaxis (ADSC-NEC, n = 66) and control group (NEC-PLCB, n = 62)-to measure the influence of ADSCs administration on the inflammatory changes in NEC, the level of cell engraftment, and the histopathology of the disease. The analysis did not show a significant effect on histopathology between groups, H(2) = 2.12; p = 0.347; η²H = 0.00. The intensity of the NEC variable results was similar across the analyzed groups (NEC-PLCB and ADSC-NEC). For IL-1 and IL-6, the difference between the NEC-PLCB group and the ADSC-NEC group was statistically significant, p = 0.002 and p < 0.001, respectively. To conclude, administration of adipose tissue-derived stem cells before the onset of the disease lowers the levels of inflammatory cytokines IL-1 and IL-6 but does not affect the histopathological results in the rat model of NEC.
Collapse
Affiliation(s)
- Marek Wolski
- Department of Pediatric Surgery, Medical University of Warsaw, Zwirki i Wigury 63a, 02-091 Warsaw, Poland
| | - Tomasz Ciesielski
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (T.C.); (K.B.); (A.C.-J.)
| | - Kasper Buczma
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (T.C.); (K.B.); (A.C.-J.)
| | - Łukasz Fus
- Department of Pathology, Medical University of Warsaw, Pawinskiego 7, 02-106 Warsaw, Poland;
| | - Agnieszka Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (A.G.); (J.T.-D.)
| | - Joanna Trzcińska-Danielewicz
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (A.G.); (J.T.-D.)
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (T.C.); (K.B.); (A.C.-J.)
| |
Collapse
|
8
|
Sarafidis K, Agakidou E, Kontou A, Agakidis C, Neu J. Struggling to Understand the NEC Spectrum-Could the Integration of Metabolomics, Clinical-Laboratory Data, and Other Emerging Technologies Help Diagnosis? Metabolites 2024; 14:521. [PMID: 39452903 PMCID: PMC11509608 DOI: 10.3390/metabo14100521] [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: 08/23/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most prevalent and potentially fatal intestinal injury mainly affecting premature infants, with significant long-term consequences for those who survive. This review explores the scale of the problem, highlighting advancements in epidemiology, the understanding of pathophysiology, and improvements in the prediction and diagnosis of this complex, multifactorial, and multifaced disease. Additionally, we focus on the potential role of metabolomics in distinguishing NEC from other conditions, which could allow for an earlier and more accurate classification of intestinal injuries in infants. By integrating metabolomic data with other diagnostic approaches, it is hoped to enhance our ability to predict outcomes and tailor treatments, ultimately improving care for affected infants.
Collapse
Affiliation(s)
- Kosmas Sarafidis
- 1st Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.A.); (A.K.)
| | - Eleni Agakidou
- 1st Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.A.); (A.K.)
| | - Angeliki Kontou
- 1st Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.A.); (A.K.)
| | - Charalampos Agakidis
- 1st Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, Gainesville, FL 32611, USA;
| |
Collapse
|
9
|
Chen J, Chen X, Huang X, Liu J, Yu Q. Comparative efficacy of different single drugs to prevent necrotizing enterocolitis in preterm infants: an update systematic review and network meta-analysis. Front Nutr 2024; 11:1452338. [PMID: 39315009 PMCID: PMC11416958 DOI: 10.3389/fnut.2024.1452338] [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: 06/20/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Objective To investigate an optimal regimen of six drugs, including lactoferrin, probiotics, prebiotics, glutamine, arginine and erythropoietin (EPO), for the prevention of necrotizing enterocolitis (NEC) in preterm infants. Methods PubMed, Embase, Ovid, The Cochrane Library, and Web of Science databases were searched for randomized controlled trials (RCTs) investigating the efficacy of lactoferrin, probiotics, prebiotics, glutamine, arginine, and EPO in preventing NEC in preterm infants, with a cutoff date of June 20, 2024. Two authors independently screened studies and extracted all the data. Network meta-analysis (NMA) was conducted to compare the outcomes of different interventions, and group rankings were determined using the surface under the cumulative ranking curve (SUCRA). Results A total of 89 RCTs with 26,861 preterm infants were included. Arginine demonstrated the highest clinical efficacy in reducing the incidence of NEC, with probiotics being the next most effective and the placebo being the least effective. Lactoferrin was identified as the most effective intervention for reducing the incidence of NEC-associated sepsis. Prebiotics showed the highest effect on overall mortality, reducing the beginning of enteral feeding, and were associated with the shortest hospital stay. Glutamine significantly decreased the time to full enteral feeding. Conclusion Existing literature highlights arginine as the most efficacious pharmacological agent in preventing NEC in preterm infants. It has been shown to effectively lower the rates of NEC, septicemia, and mortality, warranting its recommendation as the first-line clinical intervention. Following this, probiotics are recommended as a second option.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neonatology, The First People’s Hospital of Neijiang, Neijiang, China
| | - Xiao Chen
- Department of Orthopedics, The First People’s Hospital of Neijiang, Neijiang, China
| | - Xiaoling Huang
- Department of Neonatology, The First People’s Hospital of Neijiang, Neijiang, China
| | - Jia Liu
- Department of Neonatology, The First People’s Hospital of Neijiang, Neijiang, China
| | - Qingfeng Yu
- Department of Neonatology, The First People’s Hospital of Neijiang, Neijiang, China
| |
Collapse
|
10
|
Zhang Y, Tian Y, Zhong X, Zhang R, Yang S, Jin J, Lyu C, Fan J, Shi B, Zhu K, Xiao Y, Lin N, Ma D, Tou J, Shu Q, Lai D. RNF31-mediated IKKα ubiquitination aggravates inflammation and intestinal injury through regulating NF-κB activation in human and mouse neonates. Life Sci 2024; 352:122893. [PMID: 38971367 DOI: 10.1016/j.lfs.2024.122893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
AIMS Neonatal necrotizing enterocolitis (NEC) is a leading cause of intestine inflammatory disease, and macrophage is significantly activated during NEC development. Posttranslational modifications (PTMs) of proteins, particularly ubiquitination, play critical roles in immune response. This study aimed to investigate the effects of ubiquitin-modified proteins on macrophage activation and NEC, and discover novel NEC-related inflammatory proteins. MATERIALS AND METHODS Proteomic and ubiquitin proteomic analyses of intestinal macrophages in NEC/healthy mouse pups were carried out. In vitro macrophage inflammation model and in vivo NEC mouse model, as well as clinical human samples were used for further verification the inhibitor of nuclear factor-κB kinase α (IKKα) ubiquitination on NEC development through Western blot, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR) and flow cytometry. KEY FINDINGS We report here that IKKα was a new ubiquitin-modified protein during NEC through ubiquitin proteomics, and RING finger protein 31 (RNF31) acted as an E3 ligase to be involved in IKKα degradation. Inhibition of IKKα ubiquitination and degradation with siRNF31 or proteasome inhibitor decreased nuclear factor-κB (NF-κB) activation, thereby decreasing the expression of pro-inflammatory factors and M1 macrophage polarization, resulting in reliving the severity of NEC. SIGNIFICANCE Our study suggests the activation of RNF31-IKKα-NF-κB axis triggering NEC development and suppressing RNF31-mediated IKKα degradation may be therapeutic strategies to be developed for NEC treatment.
Collapse
Affiliation(s)
- Yuebai Zhang
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yangfan Tian
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaohui Zhong
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ruoyang Zhang
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingyi Jin
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chengjie Lyu
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiajie Fan
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Bo Shi
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kun Zhu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Xiao
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Nan Lin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Daqing Ma
- Perioperative and Systems Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China; Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
11
|
Shen L, Zhong X, Ji H, Yang S, Jin J, Lyu C, Ren Y, Xiao Y, Zhang Y, Fang S, Lin N, Tou J, Shu Q, Lai D. Macrophage α7nAChR alleviates the inflammation of neonatal necrotizing enterocolitis through mTOR/NLRP3/IL-1β pathway. Int Immunopharmacol 2024; 139:112590. [PMID: 38996778 DOI: 10.1016/j.intimp.2024.112590] [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: 01/18/2024] [Revised: 06/08/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Neonatal necrotizing enterocolitis (NEC) is one of the most prevalent and severe intestinal emergencies in newborns. The inflammatory activation of macrophages is associated with the intestinal injury of NEC. The neuroimmune regulation mediated by α7 nicotinic acetylcholine receptor (α7nAChR) plays an important role in regulating macrophage activation and inflammation progression, but in NEC remains unclear. This study aims to explore the effect of macrophage α7nAChR on NEC. METHODS Mice NEC model were conducted with high-osmolarity formula feeding, hypoxia, and cold stimulation. The α7nAChR agonist PNU-282987 and mTOR inhibitor rapamycin were treated by intraperitoneal injections in mice. The expression and distribution of macrophages, α7nAChR, and phospho-mammalian target of rapamycin (p-mTOR) in the intestines of NEC patients and mice was assessed using immunohistochemistry, immunofluorescence, and flow cytometry. The expression of NLRP3, activated caspase-1 and IL-1β in mice intestines was detected by flow cytometry, western blot or ELISA. In vitro, the mouse RAW264.7 macrophage cell line was also cultured followed by various treatments. Expression of p-mTOR, NLRP3, activated caspase-1, and IL-1β in macrophages was determined. RESULTS Macrophages accumulated in the intestines and the expression of α7nAChR in the mucosal and submucosal layers of the intestines was increased in both the NEC patients and mice. The p-mTOR and CD68 were increased and co-localized in intestines of NEC patients. In vitro, α7nAChR agonist PNU-282987 significantly reduced the increase of NLRP3, activated caspase-1, and IL-1β in macrophages. PNU-282987 also significantly reduced the increase of p-mTOR. The effect was blocked by AMPK inhibitor compound C. The expression of NLRP3, activated caspase-1, and IL-1β was inhibited after mTOR inhibitor rapamycin treatment. In NEC model mice, PNU-282987 reduced the expression of p-mTOR, NLRP3, activated caspase-1, and IL-1β in the intestine. Meanwhile, rapamycin significantly attenuated NLRP3 activation and the release of IL-1β. Moreover, the proportion of intestinal macrophages and intestinal injury decreased after PNU-282987 treatment. CONCLUSION Macrophage α7nAChR activation mitigates NLRP3 inflammasome activation by modulating mTOR phosphorylation, and subsequently alleviates intestinal inflammation and injury in NEC.
Collapse
Affiliation(s)
- Leiting Shen
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Xiaohui Zhong
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Haosen Ji
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jingyi Jin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Chengjie Lyu
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yichao Ren
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yi Xiao
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yuebai Zhang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Shu Fang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Nan Lin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
12
|
Korček P, Straňák Z. No differences were observed in the prevention of necrotizing enterocolitis and late-onset sepsis among preterm infants who received either single-species or multi-species probiotics. Early Hum Dev 2024; 194:106054. [PMID: 38795665 DOI: 10.1016/j.earlhumdev.2024.106054] [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: 04/17/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Probiotic prophylaxis has been suggested to reduce the incidence of necrotizing enterocolitis (NEC) and late-onset sepsis (LOS) in very preterm newborns. However, choosing the optimal probiotic is difficult due to variations in strain-specific effects and interactions facilitated by the use of combination species. AIMS To compare clinical outcomes of very preterm infants receiving multi or single-species probiotics. STUDY DESIGN Retrospective, single-center, cohort study. SUBJECTS Very preterm infants (<32 weeks' gestation) born between 2019 and 2022 at a tertiary perinatal center received either a multi-species (Lactobacillus rhamnosus 45 %, Lactobacillus casei 15 %, Lactobacillus acidophilus 15 %, Bifidobacterium infantis 15 %, Bifidobacterium bifidum 10 %; n = 228) or a single-species (Bifidobacterium breve BR03 and B632; n = 227) probiotic formulation. MAIN OUTCOME MEASURES NEC, LOS, and mortality. RESULTS The overall incidence of NEC and LOS was 3.1 % and 13.8 %, respectively. There were no differences between the multi-species and single-species probiotic groups in the rate of NEC (3.5 % vs 2.6 %; p = 0.787), LOS (15.4 % vs 12.3 %; p = 0.416), mortality (0.9 % vs 1.8 %; p = 0.449), or composite outcome (NEC, LOS and/or death; 16.7 % vs 12.8 %; p = 0.290). CONCLUSION The clinical outcomes of very preterm newborns receiving multi vs. single-species probiotic formulations were similar in our study. In view of the sample size and low baseline rate of NEC in our unit, further trials are warranted to investigate the effects of specific probiotics for prevention of serious neonatal morbidities.
Collapse
Affiliation(s)
- Peter Korček
- Institute for the Care of Mother and Child, Neonatology, Prague 147 00, Czech Republic; Third Faculty of Medicine, Charles University, Prague 100 00, Czech Republic.
| | - Zbyněk Straňák
- Institute for the Care of Mother and Child, Neonatology, Prague 147 00, Czech Republic; Third Faculty of Medicine, Charles University, Prague 100 00, Czech Republic
| |
Collapse
|
13
|
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
|
14
|
Pijpers AGH, Gorter RR, Eeftinck Schattenkerk LD, van Schuppen J, van den Akker CHP, Vanhamel S, van Heurn ELW, Musters GD, Derikx JPM. Identifying Preoperative Clinical Characteristics of Unexpected Gastrointestinal Perforation in Infants-A Retrospective Cohort Study. CHILDREN (BASEL, SWITZERLAND) 2024; 11:505. [PMID: 38790500 PMCID: PMC11119732 DOI: 10.3390/children11050505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Infants presenting with unexpected pneumoperitoneum upon abdominal X-ray, indicating a gastrointestinal perforation (GIP), have a surgical emergency with potential morbidity and mortality. Preoperative determination of the location of perforation is challenging but will aid the surgeon in optimizing the surgical strategy, as colon perforations are more challenging than small bowel perforations. Therefore, the aim of this study is to provide an overview of preoperative patient characteristics, determine the differences between the small bowel and colon, and determine underlying causes in a cohort of infants with unexpected GIP. METHODS All infants (age ≤ 6 months) who presented at our center with unexpected pneumoperitoneum (no signs of pneumatosis before) undergoing surgery between 1996 and 2024 were retrospectively included. The differences between the location of perforation were analyzed using chi-squared and t-tests. Bonferroni correction was used to adjust for multiple tests. RESULTS In total, 51 infants presented with unexpected pneumoperitoneum at our center, predominantly male (N = 36/51) and premature (N = 40/51). Among them, twenty-six had small bowel, twenty-two colon, and three stomach perforations. Prematurity (p = 0.001), birthweight < 1000 g (p = 0.001), respiratory support (p = 0.001), and lower median arterial pH levels (p = 0.001) were more present in patients with small bowel perforation compared with colon perforations. Pneumatosis intestinalis was more present in patients with colon perforation (p = 0.004). All patients with Hirschsprung disease and cystic fibrosis had colon perforation. The final diagnoses were mainly focal intestinal perforations (N = 27/51) and necrotizing enterocolitis (N = 9/51). CONCLUSIONS Infants with unexpected GIP, birthweight < 1000 g, and prematurity have more risk for small bowel perforation. In case of colon perforation, additional screening (for Hirschsprung and cystic fibrosis) should be considered.
Collapse
Affiliation(s)
- Adinda G. H. Pijpers
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Ramon R. Gorter
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Laurens D. Eeftinck Schattenkerk
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Joost van Schuppen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Chris H. P. van den Akker
- Amsterdam Reproduction & Development Research Institute, 1105 AZ Amsterdam, The Netherlands
- Department of Neonatology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sylvie Vanhamel
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ernest L. W. van Heurn
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gijsbert D. Musters
- Department of Surgery, Zaans Medisch Centrum, Koningin Julianaplein 58, 1502 DV Zaandam, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
15
|
Tuşat M, Eroz R, Bölükbaş F, Özkan E, Erdal H. Evaluation of the protective and therapeutic effects of extra virgin olive oil rich in phenol in experimental model of neonatal necrotizing enterocolitis by clinical disease score, ınflammation, apoptosis, and oxidative stress markers. Pediatr Surg Int 2024; 40:80. [PMID: 38493431 DOI: 10.1007/s00383-024-05669-1] [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] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND AIM Necrotizing Enterocolitis (NEC) is an inflammation-associated ischemic necrosis of the intestine. To investigate the effects of extra virgin olive oil (EVOO) on inflammation, oxidative stress, apoptosis, and histological changes in NEC-induced newborn rats. MATERIALS AND METHODS 24 rats were randomly divided into three groups: control, NEC and NEC + EVOO. NEC induction was performed using hypoxia-hyperoxia, formula feeding, and cold stress. The NEC + EVOO group received 2 ml/kg EVOO with high phenolic content by gavage twice a day for 3 days. 3 cm of bowel including terminal ileum, cecum, and proximal colon was excised. RESULTS Weight gain and clinical disease scores were significantly higher in the NEC + EVOO group than in the NEC group (p < 0.001). EVOO treatment caused significant decreases in IL1β, IL6 levels (p = 0.016, p = 0.029 respectively) and EGF, MDA levels (p = 0.032, p = 0.013 respectively) compared to NEC group. Significant decreases were observed in IL6 gene expression in the NEC + EVOO group compared to the NEC group (p = 0.002). In the group NEC + EVOO, the number of Caspase-3 positive cells was found to be significantly reduced (p < 0.001) and histopathological examination revealed minimal changes and significantly lower histopathological scores (p < 0.001). CONCLUSION Phenol-rich EVOO prevents intestinal damage caused by NEC by inhibiting inflammation, oxidative stress, apoptosis.
Collapse
Affiliation(s)
- Mustafa Tuşat
- Department of Pediatric Surgery, Aksaray University Medical Faculty, Aksaray, Turkey.
| | - Recep Eroz
- Department of Medical Genetics, Aksaray University Medical Faculty, Aksaray, Turkey
| | - Ferhan Bölükbaş
- Department of Histology and Embryology, Aksaray University Medical Faculty, Aksaray, Turkey
| | - Erkan Özkan
- Faculty of Veterinary Medicine, Department of Parasitology, Aksaray University, Aksaray, Turkey
| | - Hüseyin Erdal
- Department of Medical Genetics, Aksaray University Medical Faculty, Aksaray, Turkey
| |
Collapse
|
16
|
Liu S, Liu Y, Lai S, Xie Y, Xiu W, Yang C. Values of serum intestinal fatty acid-binding protein, fecal calprotectin, and fecal human β-defensin 2 for predicting necrotizing enterocolitis. BMC Pediatr 2024; 24:183. [PMID: 38491401 PMCID: PMC10943912 DOI: 10.1186/s12887-024-04667-5] [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: 12/01/2023] [Accepted: 02/23/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND This study aimed to assess the diagnostic potential of serum intestinal fatty acid-binding protein (I-FABP), fecal calprotectin (FC), and fecal human β-defensin 2 (hBD2) in predicting necrotizing enterocolitis (NEC) in preterm infants. METHODS A prospective cohort of neonates with a gestational age < 32 weeks, suspected of NEC, was enrolled between June 2021 and December 2022. Serum I-FABP, FC, and fecal hBD2 levels were measured upon NEC suspicion, and diagnosis was confirmed through radiological examination or surgical intervention. Diagnostic precision of serum I-FABP, FC, and fecal hBD2 was assessed using a logistic regression model with multiple variables. RESULTS The study included 70 neonates (45 males, 25 females), with 30 developing NEC (40% Stage III, n = 12; 60% Stage II, n = 18) and 40 in the control group. NEC patients exhibited significantly higher serum I-FABP and FC levels (4.76 ng/mL and 521.56 µg/g feces, respectively) than those with other diagnoses (1.38 ng/mL and 213.34 µg/g feces, respectively; p ˂ 0.05 for both biomarkers). Stage II NEC neonates showed elevated fecal hBD2 levels (376.44 ng/g feces) than Stage III NEC neonates and controls (336.87 ng/g and 339.86 ng/g feces, respectively; p ˂ 0.05). No such increase was observed in infants progressing to Stage III NEC. Using a serum I-FABP threshold of > 2.54 ng/mL yielded 76.7% sensitivity, 87.5% specificity, 82.1% positive predictive value (PPV), and 83.3% negative predictive value (NPV). For FC (cutoff > 428.99 µg/g feces), corresponding values were 76.7% sensitivity, 67.5% specificity, 63.9% PPV, and 79.4% NPV. CONCLUSION Serum I-FABP and FC levels are valuable for early NEC detection and provide insights into disease severity. Low fecal hBD2 levels suggest an inadequate response to luminal bacteria, potentially rendering these infants more susceptible to NEC development or exacerbation.
Collapse
Affiliation(s)
- Sujia Liu
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Fuzhou, People's Republic of China
| | - Yongle Liu
- Neonatal Intensive Care Unit, Fujian Provincial Hospital, Fuzhou, People's Republic of China
| | - Shuhua Lai
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Fuzhou, People's Republic of China
| | - Yingling Xie
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Fuzhou, People's Republic of China
| | - Wenlong Xiu
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Fuzhou, People's Republic of China
| | - Changyi Yang
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Fuzhou, People's Republic of China.
| |
Collapse
|
17
|
Opramolla A, Gazzin A, Cisarò F, Pinon M, Calvo P, Rigazio C. Intestinal ultrasonography in pediatric population. Minerva Pediatr (Torino) 2024; 76:100-107. [PMID: 33871250 DOI: 10.23736/s2724-5276.21.06371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bowel ultrasound (US) is a low-cost, non-invasive, bed side practice and a reproducible procedure that represents a high yield tool in clinical practice and in the diagnostic workup of a consistent group of acute and chronic gastrointestinal (GI) tract disease. Moreover, in case of GI diseases of neonatal and pediatric age, it allows an easier evaluation due to the small body size and scarce presence of fat tissue in the abdominal wall and peritoneal cavity and gas content. No particular preparation of the patient is needed, nevertheless a 3- to 5-hour fasting state improves the quality of the examination. The exam focuses on wall thickness and stratification, lumen content, distensibility and compressibility, presence of peristalsis of explorable segment of the GI tract and includes the investigation of mesentery, perivisceral tissues and nodes features. Color doppler flowmetry admits a qualitative evaluation of GI wall and mesentery vascularization. Healthy GI wall appears at a US evaluation as a multilayered structure in which hyperechoic and hypoechoic layers alternate sequentially. In this article we provide a quickly available overview on findings, signs and applications of US in major GI pediatric diseases.
Collapse
Affiliation(s)
- Anna Opramolla
- Unit of Pediatric Gastroenterology, Department of Pediatrics, Città della Salute e della Scienza, Turin, Italy -
| | - Andrea Gazzin
- Department of Public Health and Pediatric Sciences, School of Medicine, University of Turin, Turin, Italy
| | - Fabio Cisarò
- Unit of Pediatric Gastroenterology, Department of Pediatrics, Città della Salute e della Scienza, Turin, Italy
| | - Michele Pinon
- Unit of Pediatric Gastroenterology, Department of Pediatrics, Città della Salute e della Scienza, Turin, Italy
| | - Pierluigi Calvo
- Unit of Pediatric Gastroenterology, Department of Pediatrics, Città della Salute e della Scienza, Turin, Italy
| | - Caterina Rigazio
- Unit of Pediatric Gastroenterology, Department of Pediatrics, Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
18
|
Li Z, Yuan T. Neutrophil extracellular traps in adult diseases and neonatal bacterial infectious diseases: A review. Heliyon 2024; 10:e23559. [PMID: 38173520 PMCID: PMC10761809 DOI: 10.1016/j.heliyon.2023.e23559] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Neutrophils, the most abundant type of white blood cells, are pivotal in fighting bacterial infections due to their immunological and anti-infection capabilities. In recent years, scientists have discovered a novel mechanism known as neutrophil extracellular traps, which are fibrous networks primarily released by neutrophils that combat bacterial infections. There is a growing interest in studying NETs and their role in human infectious diseases, particularly in neonates susceptible to bacterial infections. NETs and their components have been found in various samples from neonatal-infected patients, providing a new route for early diagnosis of neonatal infectious diseases. This paper aims to summarize the studies on NETs in adult diseases and mainly discuss NETs in neonatal sepsis, necrotizing enterocolitis, and purulent meningitis, to provide scientific evidence for early monitoring, diagnosis, and treatment of neonatal infections.
Collapse
Affiliation(s)
- Ziheng Li
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| |
Collapse
|
19
|
Hu X, Liang H, Li F, Zhang R, Zhu Y, Zhu X, Xu Y. Necrotizing enterocolitis: current understanding of the prevention and management. Pediatr Surg Int 2024; 40:32. [PMID: 38196049 PMCID: PMC10776729 DOI: 10.1007/s00383-023-05619-3] [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: 12/15/2023] [Indexed: 01/11/2024]
Abstract
Necrotizing enterocolitis (NEC) is one of the diseases in neonates, with a high morbidity and mortality rate, especially in preterm infants. This review aimed to briefly introduce the latest epidemiology, susceptibility factors, and clinical diagnosis and presentation of NEC. We also organized new prevention strategies by risk factors according to different pathogeneses and then discussed new treatment methods based on Bell's staging and complications, and the classification of mild to high severity based on clinical and imaging manifestations. Such a generalization will help clinicians and researchers to gain a deeper understanding of the disease and to conduct more targeted classification, grading prevention, and exploration. We focused on prevention and treatment of the early and suspected stages of NEC, including the discovery of novel biomarkers and drugs to control disease progression. At the same time, we discussed its clinical application, future development, and shortcomings.
Collapse
Affiliation(s)
- Xiaohan Hu
- Institute of Pediatric, Children's Hospital of Soochow University, 92 Zhong Nan Street, Suzhou City, Jiangsu Province, China
- Department of Neonatology, Children's Hospital of Soochow University, 92 Zhong Nan Street, Suzhou City, Jiangsu Province, China
| | - Hansi Liang
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Fang Li
- Department of Human Anatomy and Histology and Embryology, Soochow University, Suzhou, Jiangsu Province, China
| | - Rui Zhang
- Institute of Pediatric, Children's Hospital of Soochow University, 92 Zhong Nan Street, Suzhou City, Jiangsu Province, China
| | - Yanbo Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xueping Zhu
- Institute of Pediatric, Children's Hospital of Soochow University, 92 Zhong Nan Street, Suzhou City, Jiangsu Province, China.
- Department of Neonatology, Children's Hospital of Soochow University, 92 Zhong Nan Street, Suzhou City, Jiangsu Province, China.
| | - Yunyun Xu
- Institute of Pediatric, Children's Hospital of Soochow University, 92 Zhong Nan Street, Suzhou City, Jiangsu Province, China.
| |
Collapse
|
20
|
Bosco A, Piu C, Picciau ME, Pintus R, Fanos V, Dessì A. Metabolomics in NEC: An Updated Review. Metabolites 2023; 14:14. [PMID: 38248817 PMCID: PMC10821135 DOI: 10.3390/metabo14010014] [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: 11/11/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Necrotizing enterocolitis (NEC) represents the most common and lethal acute gastrointestinal emergency of newborns, mainly affecting those born prematurely. It can lead to severe long-term sequelae and the mortality rate is approximately 25%. Furthermore, the diagnosis is difficult, especially in the early stages, due to multifactorial pathogenesis and complex clinical pictures with mild and non-specific symptoms. In addition, the existing tests have poor diagnostic value. Thus, the scientific community has been focusing its attention on the identification of non-invasive biomarkers capable of prediction, early diagnosis and discriminating NEC from other intestinal diseases in order to intervene early and block the progression of the pathology. In this regard, the use of "omics" technologies, especially metabolomics and microbiomics, could be a fundamental synergistic strategy to study the pathophysiology of NEC. In addition, a deeper knowledge of the microbiota-host cross-talk can clarify the metabolic pathways potentially involved in the pathology, allowing for the identification of specific biomarkers. In this article, the authors analyze the state-of-the-art concerning the application of metabolomics and microbiota analysis to investigate this pathology and discuss the future possibility of the metabolomic fingerprint of patients for diagnostic purposes.
Collapse
Affiliation(s)
| | | | | | | | | | - Angelica Dessì
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, 09124 Cagliari, Italy; (A.B.); (C.P.); (M.E.P.); (R.P.); (V.F.)
| |
Collapse
|
21
|
Wang Y, Florez ID, Morgan RL, Foroutan F, Chang Y, Crandon HN, Zeraatkar D, Bala MM, Mao RQ, Tao B, Shahid S, Wang X, Beyene J, Offringa M, Sherman PM, El Gouhary E, Guyatt GH, Sadeghirad B. Probiotics, Prebiotics, Lactoferrin, and Combination Products for Prevention of Mortality and Morbidity in Preterm Infants: A Systematic Review and Network Meta-Analysis. JAMA Pediatr 2023; 177:1158-1167. [PMID: 37782505 PMCID: PMC10546299 DOI: 10.1001/jamapediatrics.2023.3849] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023]
Abstract
Importance Modulation of intestinal microbiome by administering probiotics, prebiotics, or both may prevent morbidity and mortality in premature infants. Objective To assess the comparative effectiveness of alternative prophylactic strategies through a network meta-analysis (NMA) of randomized clinical trials. Data Sources MEDLINE, EMBASE, Science Citation Index Expanded, CINAHL, Scopus, Cochrane CENTRAL, and Google Scholar from inception until May 10, 2023. Study Selection Eligible trials tested probiotics, prebiotics, lactoferrin, and combination products for prevention of morbidity or mortality in preterm infants. Data Extraction and Synthesis A frequentist random-effects model was used for the NMA, and the certainty of evidence and inferences regarding relative effectiveness were assessed using the GRADE approach. Main Outcomes and Measures All-cause mortality, severe necrotizing enterocolitis, culture-proven sepsis, feeding intolerance, time to reach full enteral feeding, and duration of hospitalization. Results A total of 106 trials involving 25 840 preterm infants were included. Only multiple-strain probiotics were associated with reduced all-cause mortality compared with placebo (risk ratio [RR], 0.69; 95% CI, 0.56 to 0.86; risk difference [RD], -1.7%; 95% CI, -2.4% to -0.8%). Multiple-strain probiotics alone (vs placebo: RR, 0.38; 95% CI, 0.30 to 0.50; RD, -3.7%; 95% CI, -4.1% to -2.9%) or in combination with oligosaccharides (vs placebo: RR, 0.13; 95% CI, 0.05 to 0.37; RD, -5.1%; 95% CI, -5.6% to -3.7%) were among the most effective interventions reducing severe necrotizing enterocolitis. Single-strain probiotics in combination with lactoferrin (vs placebo RR, 0.33; 95% CI, 0.14 to 0.78; RD, -10.7%; 95% CI, -13.7% to -3.5%) were the most effective intervention for reducing sepsis. Multiple-strain probiotics alone (RR, 0.61; 95% CI, 0.46 to 0.80; RD, -10.0%; 95% CI, -13.9% to -5.1%) or in combination with oligosaccharides (RR, 0.45; 95% CI, 0.29 to 0.67; RD, -14.1%; 95% CI, -18.3% to -8.5%) and single-strain probiotics (RR, 0.61; 95% CI, 0.51 to 0.72; RD, -10.0%; 95% CI, -12.6% to -7.2%) proved of best effectiveness in reduction of feeding intolerance vs placebo. Single-strain probiotics (MD, -1.94 days; 95% CI, -2.96 to -0.92) and multistrain probiotics (MD, -2.03 days; 95% CI, -3.04 to -1.02) proved the most effective in reducing the time to reach full enteral feeding compared with placebo. Only single-strain and multistrain probiotics were associated with greater effectiveness compared with placebo in reducing duration of hospitalization (MD, -3.31 days; 95% CI, -5.05 to -1.58; and MD, -2.20 days; 95% CI, -4.08 to -0.31, respectively). Conclusions and Relevance In this systematic review and NMA, moderate- to high-certainty evidence demonstrated an association between multistrain probiotics and reduction in all-cause mortality; these interventions were also associated with the best effectiveness for other key outcomes. Combination products, including single- and multiple-strain probiotics combined with prebiotics or lactoferrin, were associated with the largest reduction in morbidity and mortality.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Ivan D. Florez
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, University of Antioquia, Medellin, Colombia
- Pediatric Intensive Care Unit, Clínica Las Americas-AUNA, Medellin, Colombia
| | - Rebecca L. Morgan
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Farid Foroutan
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Yaping Chang
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Holly N. Crandon
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Dena Zeraatkar
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
| | - Malgorzata M. Bala
- Department of Hygiene and Dietetics, Jagiellonian University Medical College, Krakow, Poland
| | - Randi Q. Mao
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Brendan Tao
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shaneela Shahid
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Xiaoqin Wang
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Joseph Beyene
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Martin Offringa
- Child Health Evaluative Sciences, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Philip M. Sherman
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Enas El Gouhary
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Gordon H. Guyatt
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Behnam Sadeghirad
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
- Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
22
|
Wei J, Meng Z, Li Z, Dang D, Wu H. New insights into intestinal macrophages in necrotizing enterocolitis: the multi-functional role and promising therapeutic application. Front Immunol 2023; 14:1261010. [PMID: 37841247 PMCID: PMC10568316 DOI: 10.3389/fimmu.2023.1261010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory intestinal disease that profoundly affects preterm infants. Currently, the pathogenesis of NEC remains controversial, resulting in limited treatment strategies. The preterm infants are thought to be susceptible to gut inflammatory disorders because of their immature immune system. In early life, intestinal macrophages (IMφs), crucial components of innate immunity, demonstrate functional plasticity and diversity in intestinal development, resistance to pathogens, maintenance of the intestinal barrier, and regulation of gut microbiota. When the stimulations of environmental, dietary, and bacterial factors interrupt the homeostatic processes of IMφs, they will lead to intestinal disease, such as NEC. This review focuses on the IMφs related pathogenesis in NEC, discusses the multi-functional roles and relevant molecular mechanisms of IMφs in preterm infants, and explores promising therapeutic application for NEC.
Collapse
Affiliation(s)
- Jiaqi Wei
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| | - Zhaoli Meng
- Department of Translational Medicine Research Institute, First Hospital of Jilin University, Changchun, China
| | - Zhenyu Li
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| | - Dan Dang
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| | - Hui Wu
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
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
|
24
|
Shen Y, Lin Y, Fang YF, Wu DM, He YB. Efficacy of peritoneal drainage in very-low-birth-weight neonates with Bell's stage II necrotizing enterocolitis: A single-center retrospective study. World J Gastrointest Surg 2023; 15:1416-1422. [PMID: 37555126 PMCID: PMC10405105 DOI: 10.4240/wjgs.v15.i7.1416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Currently, pediatric surgeons are challenged by a lack of consensus on the optimal management strategy (conservative or surgical) for children with Bell's stage II necrotizing enterocolitis (NEC). AIM To evaluate the clinical efficacy of peritoneal drainage in very-low-birth-weight (VLBW) neonates with modified Bell's stage II NEC. METHODS This was a retrospective analysis of 102 NEC (modified Bell's stage II) neonates born with VLBW who were treated at the Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center) between January 2017 and January 2020; these included 24 cases in the peritoneal drainage group, 36 cases in the exploratory laparotomy group, and 42 cases in the conservative treatment group. RESULTS The general characteristics were comparable in the three groups (P > 0.05). Compared with conservative treatment, peritoneal drainage was associated with significantly shorter fasting time, abdominal distension relief time, fecal occult blood (OB) negative conversion time, and reduced hospital length of stay (HLOS) (P < 0.05 for all). Despite some advantages of peritoneal drainage over conservative treatment in terms of cure, conversion to laparotomy, intestinal perforation, intestinal stenosis, and abdominal abscess rates, the differences were not statistically significant (P > 0.05). Compared to exploratory laparotomy, the fecal OB negative conversion time was significantly shorter in the peritoneal drainage group (P < 0.05); similarly, the exploratory laparotomy group showed longer fasting time, abdominal distension relief time, HLOS, and higher complication rate compared to peritoneal drainage group, but the between-group differences were not statistically significant (P > 0.05). CONCLUSION Peritoneal drainage, an easy-to-operate procedure, can improve the clinical symptoms of VLBW neonates with Bell's stage II NEC and help reduce the HLOS.
Collapse
Affiliation(s)
- Yong Shen
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350000, Fujian Province, China
| | - Yu Lin
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350000, Fujian Province, China
| | - Yi-Fan Fang
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350000, Fujian Province, China
| | - Dian-Ming Wu
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350000, Fujian Province, China
| | - Yuan-Bin He
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350000, Fujian Province, China
| |
Collapse
|
25
|
Sharif S, Meader N, Oddie SJ, Rojas-Reyes MX, McGuire W. Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2023; 7:CD005496. [PMID: 37493095 PMCID: PMC10370900 DOI: 10.1002/14651858.cd005496.pub6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
BACKGROUND Intestinal dysbiosis may contribute to the pathogenesis of necrotising enterocolitis (NEC) in very preterm or very low birth weight (VLBW) infants. Dietary supplementation with probiotics to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of NEC and associated mortality and morbidity in very preterm or VLBW infants. OBJECTIVES To determine the effect of supplemental probiotics on the risk of NEC and associated mortality and morbidity in very preterm or very low birth weight infants. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, the Maternity and Infant Care database, and CINAHL from inception to July 2022. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing probiotics with placebo or no probiotics in very preterm infants (born before 32 weeks' gestation) and VLBW infants (weighing less than 1500 g at birth). DATA COLLECTION AND ANALYSIS Two review authors independently evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratios (RRs), risk differences (RDs), and mean differences (MDs), with associated 95% confidence intervals (CIs). The primary outcomes were NEC and all-cause mortality; secondary outcome measures were late-onset invasive infection (more than 48 hours after birth), duration of hospitalisation from birth, and neurodevelopmental impairment. We used the GRADE approach to assess the certainty of the evidence. MAIN RESULTS We included 60 trials with 11,156 infants. Most trials were small (median sample size 145 infants). The main potential sources of bias were unclear reporting of methods for concealing allocation and masking caregivers or investigators in about half of the trials. The formulation of the probiotics varied across trials. The most common preparations contained Bifidobacterium spp., Lactobacillus spp., Saccharomyces spp., andStreptococcus spp., alone or in combination. Very preterm or very low birth weight infants Probiotics may reduce the risk of NEC (RR 0.54, 95% CI 0.46 to 0.65; I² = 17%; 57 trials, 10,918 infants; low certainty). The number needed to treat for an additional beneficial outcome (NNTB) was 33 (95% CI 25 to 50). Probiotics probably reduce mortality slightly (RR 0.77, 95% CI 0.66 to 0.90; I² = 0%; 54 trials, 10,484 infants; moderate certainty); the NNTB was 50 (95% CI 50 to 100). Probiotics probably have little or no effect on the risk of late-onset invasive infection (RR 0.89, 95% CI 0.82 to 0.97; I² = 22%; 49 trials, 9876 infants; moderate certainty). Probiotics may have little or no effect on neurodevelopmental impairment (RR 1.03, 95% CI 0.84 to 1.26; I² = 0%; 5 trials, 1518 infants; low certainty). Extremely preterm or extremely low birth weight infants Few data were available for extremely preterm or extremely low birth weight (ELBW) infants. In this population, probiotics may have little or no effect on NEC (RR 0.92, 95% CI 0.69 to 1.22, I² = 0%; 10 trials, 1836 infants; low certainty), all-cause mortality (RR 0.92, 95% CI 0.72 to 1.18; I² = 0%; 7 trials, 1723 infants; low certainty), or late-onset invasive infection (RR 0.93, 95% CI 0.78 to 1.09; I² = 0%; 7 trials, 1533 infants; low certainty). No trials provided data for measures of neurodevelopmental impairment in extremely preterm or ELBW infants. AUTHORS' CONCLUSIONS Given the low to moderate certainty of evidence for the effects of probiotic supplements on the risk of NEC and associated morbidity and mortality for very preterm or VLBW infants, and particularly for extremely preterm or ELBW infants, there is a need for further large, high-quality trials to provide evidence of sufficient validity and applicability to inform policy and practice.
Collapse
Key Words
- female
- humans
- infant
- infant, newborn
- enterocolitis, necrotizing
- enterocolitis, necrotizing/epidemiology
- fetal growth retardation
- infant, extremely premature
- infant, premature, diseases
- infant, premature, diseases/etiology
- infant, premature, diseases/prevention & control
- infant, very low birth weight
- probiotics
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Nicholas Meader
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam J Oddie
- Centre for Reviews and Dissemination, University of York, York, UK
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Maria X Rojas-Reyes
- Institut d'Recerca Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Evaluation Unit of the Canary Islands Health Service (SESCS), Tenerife, Spain
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
26
|
Yang S, Wei X, Zhong Y, Guo C, Liu X, Wang Z, Tu Y. Programmed death of intestinal epithelial cells in neonatal necrotizing enterocolitis: a mini-review. Front Pediatr 2023; 11:1199878. [PMID: 37342533 PMCID: PMC10277470 DOI: 10.3389/fped.2023.1199878] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is one of the most fatal diseases in premature infants. Damage to the intestinal epithelial barrier (IEB) is an important event in the development of intestinal inflammation and the evolution of NEC. The intestinal epithelial monolayer formed by the tight arrangement of intestinal epithelial cells (IECs) constitutes the functional IEB between the organism and the extra-intestinal environment. Programmed death and regenerative repair of IECs are important physiological processes to maintain the integrity of IEB function in response to microbial invasion. However, excessive programmed death of IECs leads to increased intestinal permeability and IEB dysfunction. Therefore, one of the most fundamental questions in the field of NEC research is to reveal the pathological death process of IECs, which is essential to clarify the pathogenesis of NEC. This review focuses on the currently known death modes of IECs in NEC mainly including apoptosis, necroptosis, pyroptosis, ferroptosis, and abnormal autophagy. Furthermore, we elaborate on the prospect of targeting IECs death as a treatment for NEC based on exciting animal and clinical studies.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Xin Wei
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuting Zhong
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Conglu Guo
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Xinzhu Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhibin Wang
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Ye Tu
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Sharif S, Oddie SJ, Heath PT, McGuire W. Prebiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2023; 6:CD015133. [PMID: 37262358 PMCID: PMC10234253 DOI: 10.1002/14651858.cd015133.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
BACKGROUND Dietary supplementation with prebiotic oligosaccharides to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of necrotising enterocolitis (NEC) and associated mortality and morbidity in very preterm or very low birth weight (VLBW) infants. OBJECTIVES To assess the benefits and harms of enteral supplementation with prebiotics (versus placebo or no treatment) for preventing NEC and associated morbidity and mortality in very preterm or VLBW infants. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, the Maternity and Infant Care database and the Cumulative Index to Nursing and Allied Health Literature (CINAHL), from the earliest records to July 2022. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing prebiotics with placebo or no prebiotics in very preterm (< 32 weeks' gestation) or VLBW (< 1500 g) infants. The primary outcomes were NEC and all-cause mortality, and the secondary outcomes were late-onset invasive infection, duration of hospitalisation since birth, and neurodevelopmental impairment. DATA COLLECTION AND ANALYSIS Two review authors separately evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference (MD), with associated 95% confidence intervals (CIs). The primary outcomes of interest were NEC and all-cause mortality; our secondary outcome measures were late-onset (> 48 hours after birth) invasive infection, duration of hospitalisation, and neurodevelopmental impairment. We used the GRADE approach to assess the level of certainty of the evidence. MAIN RESULTS We included seven trials in which a total of 705 infants participated. All the trials were small (mean sample size 100). Lack of clarity on methods to conceal allocation and mask caregivers or investigators were potential sources of bias in three of the trials. The studied prebiotics were fructo- and galacto-oligosaccharides, inulin, and lactulose, typically administered daily with enteral feeds during birth hospitalisation. Meta-analyses of data from seven trials (686 infants) suggest that prebiotics may result in little or no difference in NEC (RR 0.97, 95% CI 0.60 to 1.56; RD none fewer per 1000, 95% CI 50 fewer to 40 more; low-certainty evidence), all-cause mortality (RR 0.43, 95% CI 0.20 to 0.92; 40 per 1000 fewer, 95% CI 70 fewer to none fewer; low-certainty evidence), or late-onset invasive infection (RR 0.79, 95% CI 0.60 to 1.06; 50 per 1000 fewer, 95% CI 100 fewer to 10 more; low-certainty evidence) prior to hospital discharge. The certainty of this evidence is low because of concerns about the risk of bias in some trials and the imprecision of the effect size estimates. The data available from one trial provided only very low-certainty evidence about the effect of prebiotics on measures of neurodevelopmental impairment (Bayley Scales of Infant Development (BSID) Mental Development Index score < 85: RR 0.84, 95% CI 0.25 to 2.90; very low-certainty evidence; BSID Psychomotor Development Index score < 85: RR 0.24, 95% 0.03 to 2.00; very low-certainty evidence; cerebral palsy: RR 0.35, 95% CI 0.01 to 8.35; very low-certainty evidence). AUTHORS' CONCLUSIONS The available trial data provide low-certainty evidence about the effects of prebiotics on the risk of NEC, all-cause mortality before discharge, and invasive infection, and very low-certainty evidence about the effect on neurodevelopmental impairment for very preterm or VLBW infants. Our confidence in the effect estimates is limited; the true effects may be substantially different. Large, high-quality trials are needed to provide evidence of sufficient validity to inform policy and practice decisions.
Collapse
Key Words
- humans
- infant, newborn
- enterocolitis, necrotizing
- enterocolitis, necrotizing/etiology
- enterocolitis, necrotizing/prevention & control
- infant, extremely premature
- infant, premature, diseases
- infant, premature, diseases/etiology
- infant, premature, diseases/prevention & control
- infant, very low birth weight
- infections
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute, St. George's, University of London, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
28
|
Mi Y, Xie X, Bao Z, Xiong X, Wang X, Zhang H. Dimethyl fumarate protects against intestine damage in necrotizing enterocolitis by inhibiting the Toll-like receptor (TLR) inflammatory signaling pathway. Tissue Cell 2023; 81:102003. [PMID: 36682224 DOI: 10.1016/j.tice.2022.102003] [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: 09/16/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Necrotizing enterocolitis (NEC) is a severe disease in newborns, this study aimed to investigate the protective effect of dimethyl fumarate (DMF) on NEC and its possible mechanism. METHODS In vivo, the mice were divided into the control, NEC, and NEC+DMF group. The NEC model was established by artificial feeding, hypoxic for 4 days, and lipopolysaccharide (LPS) stimulation on day 2 and day 3. DMF (25 mg/kg/d) was administered to NEC mice on day 1 and day 3. On the 11th day, the blood and intestinal tissues of mice were taken for enzyme-linked immunosorbent assay (ELISA), pathological examination, quantitative real-time PCR (RT-qPCR), Western blot, and immunohistochemical (IHC) detection. In vitro, human colorectal cells (FHC) were induced by LPS (100 ng/mL) and was divided into the control, LPS, and LPS+DMF group. The effect of DMF (20 μM) on cell viability and TLR4 signal transduction was detected by MTT and RT-qPCR, respectively. RESULTS Compared to the NEC mice, DMF attenuated NEC-induced weight loss and abdominal distension diarrhea in mice, and alleviated NEC-induced intestinal pathological injuries. In addition, DMF reduced the expression of IL-6, IL-1β, TNF-α, NF-κB, and TLR4 in NEC mice intestinal tissues. Furthermore, DMF inhibited NEC-induced intestinal cell apoptosis as well as the protein expression of BCL2-Associated X (BAX), caspase-3, caspase-9, and increased Bcl-2 (B-cell lymphoma-2) expression. In vitro, DMF improved cell viability, and restrained NF-κB and TLR4 expression in LPS-induced NEC cells. CONCLUSION DMF has a protective effect against intestine damage of NEC, which is related to the inhibition of the TLR signaling pathway, alleviating the inflammatory response.
Collapse
Affiliation(s)
- Yanhong Mi
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou, Zhejiang Province, China
| | - Xiaoxiao Xie
- Department of Radiology, Wu YunShan Hospital of Hangzhou, Hangzhou, Zhejiang Province, China
| | - Zhongkun Bao
- Department of Radiology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xiaoyu Xiong
- Department of Neonatology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xinhong Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Hongxi Zhang
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
29
|
Lee ES, Kim EK, Shin SH, Jung YH, Song IG, Kim YJ, Kim HY, Choi YH, Moon KC, Kim B. Efficacy and safety of mucous fistula refeeding in preterm infants: an exploratory randomized controlled trial. BMC Pediatr 2023; 23:137. [PMID: 36991415 PMCID: PMC10053085 DOI: 10.1186/s12887-023-03950-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND This study aimed to evaluate whether mucous fistula refeeding (MFR) is safe and beneficial for the growth and intestinal adaptation of preterm infants with enterostomies. METHODS This exploratory randomized controlled trial enrolled infants born before 35 weeks' gestation with enterostomy. If the stomal output was ≥ 40 mL/kg/day, infants were assigned to the high-output MFR group and received MFR. If the stoma output was < 40 mL/kg/day, infants were randomized to the normal-output MFR group or the control group. Growth, serum citrulline levels, and bowel diameter in loopograms were compared. The safety of MFR was evaluated. RESULTS Twenty infants were included. The growth rate increased considerably, and the colon diameter was significantly larger after MFR. However, the citrulline levels did not significantly differ between the normal-output MFR and the control group. One case of bowel perforation occurred during the manual reduction for stoma prolapse. Although the association with MFR was unclear, two cases of culture-proven sepsis during MFR were noted. CONCLUSIONS MFR benefits the growth and intestinal adaptation of preterm infants with enterostomy and can be safely implemented with a standardized protocol. However, infectious complications need to be investigated further. TRIAL REGISTRATION clinicaltrials.gov NCT02812095, retrospectively registered on June 6, 2016.
Collapse
Affiliation(s)
- Eun Sun Lee
- Department of Pediatrics, Seoul National University Children's Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, South Korea
- Department of Pediatrics, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Ee-Kyung Kim
- Department of Pediatrics, Seoul National University Children's Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, South Korea.
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Seung Han Shin
- Department of Pediatrics, Seoul National University Children's Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, South Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Hwa Jung
- Department of Pediatrics, Seoul National University Children's Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, South Korea
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In-Gyu Song
- Department of Pediatrics, Seoul National University Children's Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, South Korea
- Department of Pediatrics, Korea University Medical Centre, Guro Hospital, Seoul, Republic of Korea
| | - Yoo-Jin Kim
- Department of Pediatrics, Seoul National University Children's Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, South Korea
- Department of Pediatrics, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Hyun Young Kim
- Department of Pediatric Surgery, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Young-Hun Choi
- Department of Radiology, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Bohyun Kim
- Department of Pathology, Seoul National University Children's Hospital, Seoul, Republic of Korea
| |
Collapse
|
30
|
Li W, Zhang C, Li W, Qin F, Gao X, Xu F. Nomogram for predicting fulminant necrotizing enterocolitis. Pediatr Surg Int 2023; 39:154. [PMID: 36939896 PMCID: PMC10027821 DOI: 10.1007/s00383-023-05435-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND Fulminant necrotizing enterocolitis (FNEC) is the most serious subtype of NEC and has a high mortality rate and a high incidence of sequelae. Onset prediction can help in the establishment of a customized treatment strategy. This study aimed to develop and evaluate a predictive nomogram for FNEC. METHODS We conducted a retrospective observation to study the clinical data of neonates diagnosed with NEC (Bell stage ≥ IIB). Neonates were divided into the FNEC and NEC groups. A multivariate logistic regression model was used to construct the nomogram model. The performance of the nomogram was assessed using area under the curve, calibration analysis, and decision curve analysis. RESULTS A total of 206 neonate cases were included, among which 40 (19.4%) fulfilled the definition of FNEC. The identified predictors were assisted ventilation after NEC onset; shock at NEC onset; feeding volumes before NEC onset; neutrophil counts on the day of NEC onset; and neutrophil, lymphocyte, and monocyte counts on day 1 after NEC onset. The nomogram exhibited good discrimination, with an area under the receiver operating characteristic curve of 0.884 (95% CI 0.825-0.943). The predictive model was well calibrated. Decision curve analysis confirmed the clinical usefulness of this nomogram. CONCLUSION A nomogram with a potentially effective application was developed to facilitate the individualized prediction of FNEC, with the hope of providing further direction for the early diagnosis of FNEC and timing of intervention.
Collapse
Affiliation(s)
- Weibo Li
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Chen Zhang
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenli Li
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Fanyue Qin
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang Gao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Falin Xu
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
31
|
Wang K, Tao GZ, Salimi-Jazi F, Lin PY, Sun Z, Liu B, Sinclair T, Mostaghimi M, Dunn J, Sylvester KG. Butyrate induces development-dependent necrotizing enterocolitis-like intestinal epithelial injury via necroptosis. Pediatr Res 2023; 93:801-809. [PMID: 36202969 DOI: 10.1038/s41390-022-02333-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/12/2022] [Accepted: 09/18/2022] [Indexed: 03/05/2023]
Abstract
BACKGROUND The accumulation of short-chain fatty acids (SCFAs) from bacterial fermentation may adversely affect the under-developed gut as observed in premature newborns at risk for necrotizing enterocolitis (NEC). This study explores the mechanism by which specific SCFA fermentation products may injure the premature newborn intestine mucosa leading to NEC-like intestinal cell injury. METHODS Intraluminal injections of sodium butyrate were administered to 14- and 28-day-old mice, whose small intestine and stool were harvested for analysis. Human intestinal epithelial stem cells (hIESCs) and differentiated enterocytes from preterm and term infants were treated with sodium butyrate at varying concentrations. Necrosulfonamide (NSA) and necrostatin-1 (Nec-1) were used to determine the protective effects of necroptosis inhibitors on butyrate-induced cell injury. RESULTS The more severe intestinal epithelial injury was observed in younger mice upon exposure to butyrate (p = 0.02). Enterocytes from preterm newborns demonstrated a significant increase in sensitivity to butyrate-induced cell injury compared to term newborn enterocytes (p = 0.068, hIESCs; p = 0.038, differentiated cells). NSA and Nec-1 significantly inhibited the cell death induced by butyrate. CONCLUSIONS Butyrate induces developmental stage-dependent intestinal injury that resembles NEC. A primary mechanism of cell injury in NEC is necroptosis. Necroptosis inhibition may represent a potential preventive or therapeutic strategy for NEC. IMPACT Butyrate induces developmental stage-dependent intestinal injury that resembles NEC. A primary mechanism of cell injury caused by butyrate in NEC is necroptosis. Necroptosis inhibitors proved effective at significantly ameliorating the enteral toxicity of butyrate and thereby suggest a novel mechanism and approach to the prevention and treatment of NEC in premature newborns.
Collapse
Affiliation(s)
- Kewei Wang
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, 110001, Shenyang, Liaoning Province, China
| | - Guo-Zhong Tao
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| | | | - Po-Yu Lin
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhen Sun
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo Liu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tiffany Sinclair
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mirko Mostaghimi
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - James Dunn
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl G Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Metabolic Health Center, Stanford University School of Medicine and Stanford Healthcare, Stanford, CA, USA.
| |
Collapse
|
32
|
Current pain management practices for preterm infants with necrotizing enterocolitis: a European survey. Pediatr Res 2023:10.1038/s41390-023-02508-2. [PMID: 36828969 PMCID: PMC10382315 DOI: 10.1038/s41390-023-02508-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a highly painful intestinal complication in preterm infants that requires adequate pain management to prevent short- and long-term effects of neonatal pain. There is a lack of international guidelines for pain management in NEC patients. Therefore, this study aims to describe current pain management for NEC patients in European neonatal intensive care units (NICUs). METHODS An online survey was designed and conducted to assess current practices in pain management for NEC patients in European NICUs. The survey was distributed via neonatal societies, digital platforms, and professional contacts. RESULTS Out of the 259 responding unique European NICUs from 36 countries, 61% had a standard protocol for analgesic therapy, 73% assessed pain during NEC, and 92% treated NEC patients with intravenous analgosedatives. There was strong heterogeneity in the used pain scales and initial analgesic therapy, which mainly included acetaminophen (70%), fentanyl (56%), and/or morphine (49%). A third of NICU representatives considered their pain assessment adequate, and half considered their analgesic therapy adequate for NEC patients. CONCLUSIONS Various pain scales and analgesics are used to treat NEC patients in European NICUs. Our results provide the first step towards an international guideline to improve pain management for NEC patients. IMPACT This study provides an overview of current pain management practices for infants with necrotizing enterocolitis (NEC) in European neonatal intensive care units. Choice of pain assessment tools, analgosedatives, and dosages vary considerably among NICUs and countries. A third of NICU representatives were satisfied with their current pain assessment practices and half of NICU representatives with their analgesic therapy practices in NEC patients in their NICU. The results of this survey may provide a first step towards developing a European pain management consensus guideline for patients with NEC.
Collapse
|
33
|
Wang H, Meng X, Xing S, Guo B, Chen Y, Pan YQ. Probiotics to prevent necrotizing enterocolitis and reduce mortality in neonates: A meta-analysis. Medicine (Baltimore) 2023; 102:e32932. [PMID: 36827026 PMCID: PMC11309691 DOI: 10.1097/md.0000000000032932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Probiotics are gradually being used as a supplementation to prevent necrotizing enterocolitis (NEC) and reduce mortality in neonates. We performed an updated meta-analysis to systematically evaluate the efficacy and safety of prophylactic probiotic supplementation for preventing NEC. METHODS The databases including PubMed, Embase, Scopus, Web of Science, and China National Knowledge Infrastructure were used to search the relevant articles. The latest retrieval date was up to December 2021. The meta-analysis was performed using Stata version 10.0. Finally, a total of 70 studies containing 8319 cases and 9283 controls were included. The strength of the association between the supplementation of probiotics and NEC was measured by risk ratios (RRs) with 95% confidence intervals (CIs). Pooled effect sizes across studies were performed by a random effect model. RESULTS The results showed that the probiotics could significantly reduce the incidence of NEC (stage II or more) (RR = 0.436, 95% CI = 0.357-0.531, P < .001), the overall mortality (RR = 0.651, 95% CI = 0.506-0.836, P < .001), and NEC-related mortality (RR = 0.639, 95% CI = 0.423-0.966, P = .034). Due to the lack of sufficient sample size, we did not perform the subgroup analysis by types of probiotic strain. CONCLUSION This meta-analysis indicates that the use of probiotics can effectively reduce the occurrence of NEC and mortality in neonates.
Collapse
Affiliation(s)
- Hongbo Wang
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Xinyao Meng
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Shihan Xing
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Baotong Guo
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Yuhan Chen
- Department of Anatomy, Shenyang Medical College, Shenyang, China
| | - Yu-Qing Pan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|
34
|
Duess JW, Sampah ME, Lopez CM, Tsuboi K, Scheese DJ, Sodhi CP, Hackam DJ. Necrotizing enterocolitis, gut microbes, and sepsis. Gut Microbes 2023; 15:2221470. [PMID: 37312412 PMCID: PMC10269420 DOI: 10.1080/19490976.2023.2221470] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease in premature infants and the leading cause of death and disability from gastrointestinal disease in this vulnerable population. Although the pathophysiology of NEC remains incompletely understood, current thinking indicates that the disease develops in response to dietary and bacterial factors in the setting of a vulnerable host. As NEC progresses, intestinal perforation can result in serious infection with the development of overwhelming sepsis. In seeking to understand the mechanisms by which bacterial signaling on the intestinal epithelium can lead to NEC, we have shown that the gram-negative bacterial receptor toll-like receptor 4 is a critical regulator of NEC development, a finding that has been confirmed by many other groups. This review article provides recent findings on the interaction of microbial signaling, the immature immune system, intestinal ischemia, and systemic inflammation in the pathogenesis of NEC and the development of sepsis. We will also review promising therapeutic approaches that show efficacy in pre-clinical studies.
Collapse
Affiliation(s)
- Johannes W. Duess
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Maame E. Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Carla M. Lopez
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Koichi Tsuboi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Daniel J. Scheese
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Chhinder P. Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
35
|
Metzler M, Burns W, Mitchell C, Napolitano S, Chaudhari BP. A case report of necrotizing enterocolitis in a moderately preterm neonate with LCHADD-A call to focus on the basics while utilizing advanced new therapies. Front Pediatr 2023; 11:1081802. [PMID: 36861082 PMCID: PMC9969157 DOI: 10.3389/fped.2023.1081802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/16/2023] [Indexed: 02/15/2023] Open
Abstract
Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is an autosomal recessive condition of impaired beta-oxidation. Traditionally, treatment included restriction of dietary long-chain fatty acids via a low-fat diet and supplementation of medium chain triglycerides. In 2020, triheptanoin received FDA approval as an alternative source of medium chain fatty acids for individuals with long-chain fatty acid oxidation disorders (LC-FAOD). We present a case of a moderately preterm neonate born at 33 2/7 weeks gestational age with LCHADD who received triheptanoin and developed necrotizing enterocolitis (NEC). Prematurity is known as a major risk factor for NEC, with risk increasing with decreasing gestational age. To our knowledge, NEC has not previously been reported in patients with LCHADD or with triheptanoin use. While metabolic formula is part of the standard of care for LC-FAOD in early life, preterm neonates may benefit from more aggressive attempts to use skimmed human milk to minimize exposure to formula during the risk period for NEC during feed advancement. This risk period may be longer in neonates with LC-FAOD compared to otherwise healthy premature neonates.
Collapse
Affiliation(s)
- Marina Metzler
- Pediatric Residency, Nationwide Children's Hospital, Columbus, OH, United States
| | - William Burns
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Carly Mitchell
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Stephanie Napolitano
- Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, United States
| | - Bimal P Chaudhari
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, United States.,Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
36
|
Kolba N, Cheng J, Jackson CD, Tako E. Intra-Amniotic Administration-An Emerging Method to Investigate Necrotizing Enterocolitis, In Vivo ( Gallus gallus). Nutrients 2022; 14:nu14224795. [PMID: 36432481 PMCID: PMC9696943 DOI: 10.3390/nu14224795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease in premature infants and a leading cause of death in neonates (1-7% in the US). NEC is caused by opportunistic bacteria, which cause gut dysbiosis and inflammation and ultimately result in intestinal necrosis. Previous studies have utilized the rodent and pig models to mimic NEC, whereas the current study uses the in vivo (Gallus gallus) intra-amniotic administration approach to investigate NEC. On incubation day 17, broiler chicken (Gallus gallus) viable embryos were injected intra-amniotically with 1 mL dextran sodium sulfate (DSS) in H2O. Four treatment groups (0.1%, 0.25%, 0.5%, and 0.75% DSS) and two controls (H2O/non-injected controls) were administered. We observed a significant increase in intestinal permeability and negative intestinal morphological changes, specifically, decreased villus surface area and goblet cell diameter in the 0.50% and 0.75% DSS groups. Furthermore, there was a significant increase in pathogenic bacterial (E. coli spp. and Klebsiella spp.) abundances in the 0.75% DSS group compared to the control groups, demonstrating cecal microbiota dysbiosis. These results demonstrate significant physiopathology of NEC and negative bacterial-host interactions within a premature gastrointestinal system. Our present study demonstrates a novel model of NEC through intra-amniotic administration to study the effects of NEC on intestinal functionality, morphology, and gut microbiota in vivo.
Collapse
Affiliation(s)
| | | | | | - Elad Tako
- Correspondence: ; Tel.: +1-607-255-0884
| |
Collapse
|
37
|
Huang D, Wang P, Chen J, Li Y, Zhu M, Tang Y, Zhou W. Selective targeting of MD2 attenuates intestinal inflammation and prevents neonatal necrotizing enterocolitis by suppressing TLR4 signaling. Front Immunol 2022; 13:995791. [PMID: 36389716 PMCID: PMC9663461 DOI: 10.3389/fimmu.2022.995791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/14/2022] [Indexed: 10/17/2023] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is an inflammatory disease that occurs in premature infants and has a high mortality rate; however, the mechanisms behind this disease remain unclear. The TLR4 signaling pathway in intestinal epithelial cells, mediated by TLR4, is important for the activation of the inflammatory storm in NEC infants. Myeloid differentiation protein 2 (MD2) is a key auxiliary component of the TLR4 signaling pathway. In this study, MD2 was found to be significantly increased in intestinal tissues of NEC patients at the acute stage. We further confirmed that MD2 was upregulated in NEC rats. MD2 inhibitor (MI) pretreatment reduced the occurrence and severity of NEC in neonatal rats, inhibited the activation of NF-κB and the release of inflammatory molecules (TNF-α and IL-6), and reduced the severity of intestinal injury. MI pretreatment significantly reduced enterocyte apoptosis while also maintaining tight junction proteins, including occludin and claudin-1, and protecting intestinal mucosal permeability in NEC rats. In addition, an NEC in vitro model was established by stimulating IEC-6 enterocytes with LPS. MD2 overexpression in IEC-6 enterocytes significantly activated NF-κB. Further, both MD2 silencing and MI pretreatment inhibited the inflammatory response. Overexpression of MD2 increased damage to the IEC-6 monolayer cell barrier, while both MD2 silencing and MI pretreatment played a protective role. In conclusion, MD2 triggers an inflammatory response through the TLR4 signaling pathway, leading to intestinal mucosal injury in NEC. In addition, MI alleviates inflammation and reduces intestinal mucosal injury caused by the inflammatory response by blocking the TLR4-MD2/NF-κB signaling axis. These results suggest that inhibiting MD2 may be an important way to prevent NEC.
Collapse
Affiliation(s)
- Dabin Huang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ping Wang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Juncao Chen
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanbin Li
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaping Tang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhou
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
Wang X, Sadeghirad B, Morgan RL, Zeratkaar D, Chang Y, Crandon HN, Couban R, Foroutan F, Florez ID. Amino acids for the prevention of mortality and morbidity in preterm infants: a systematic review and network meta-analysis. Sci Rep 2022; 12:18333. [PMID: 36316436 PMCID: PMC9622873 DOI: 10.1038/s41598-022-21318-w] [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] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
To determine the effectiveness and safety of amino acids in preventing the mortality and morbidity among preterm infants. We conducted a systematic review and network meta-analysis. We searched MEDLINE, EMBASE, Web of Science, CINAHL, Scopus, Cochrane, and Google Scholar, and grey literature, from databases inception to January 2021. We included randomized trials that evaluated any amino acids on preterm or low-birth weight infants. We performed frequentist pairwise and network meta-analyses and used the GRADE methodology to assess the certainty of the evidence and provide a summary of the results.We included 18 trials (3702 infants). Low certainty evidence showed that there seems to be no benefit for arginine, glutamine, or N-acetylcysteine in reducing all-cause mortality. Oral arginine likely results in reduction of necrotizin enterocolitis (NEC) stage ≥ II (OR 0.48; 95% CI 0.26-0.90; moderate certainty). Oral glutamine may reduce the likelihood of developing late-onset sepsis (LOS) compared to placebo (OR 0.62; 95% CI 0.47-0.82; low certainty); and likely reduces time to reach full enteral feeding (MD = - 2.63 days; 95% CI - 4.99 to - 0.27; moderate certainty). Amino acids may have no effect on mortality. Oral arginine may reduce severe NEC, and oral glutamine may reduce LOS and the time to reach full feeding.Systematic review registration: PROSPERO registration number: CRD4201603873.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Canada
| | - Behnam Sadeghirad
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Canada
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Canada
- Department of Anesthesia, McMaster University, Hamilton, ON, Canada
| | - Rebecca L Morgan
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Canada
| | - Dena Zeratkaar
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Canada
| | - Yaping Chang
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Canada
- OrthoEvidence, Burlington, Canada
| | - Holly N Crandon
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Rachel Couban
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Canada
| | - Farid Foroutan
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Canada
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre, Toronto, Canada
| | - Ivan D Florez
- Department of Pediatrics, University of Antioquia, Calle 67 No. 53-108, Medellin, Colombia.
- School of Rehabilitation Science, McMaster University, Hamilton, Canada.
- Pediatric Intensive Care Unit, Clínica Las Americas, Medellin, Colombia.
| |
Collapse
|
39
|
Mutanen A, Pöntinen V, Gunnar R, Merras-Salmio L, Pakarinen MP. Efficient achievement of enteral autonomy facilitates resolution of liver injury in necrotizing enterocolitis induced short bowel syndrome. Sci Rep 2022; 12:17516. [PMID: 36266329 PMCID: PMC9584958 DOI: 10.1038/s41598-022-22414-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/14/2022] [Indexed: 01/12/2023] Open
Abstract
Children with short bowel syndrome (SBS) are at high risk for intestinal failure associated liver disease (IFALD). The aim of this retrospective follow-up study was to compare weaning off parenteral nutrition (PN) and IFALD between necrotizing enterocolitis (NEC) and non-NEC induced SBS. Altogether, 77 patients with neonatal SBS treated by our multidisciplinary intestinal failure unit (NEC n = 38, non-NEC SBS n = 39) were included and followed-up at least for 2 years until median age of 10 years (interquartile range, 6.0-16). Occurrence and characteristics of IFALD was assessed with liver biopsies obtained at median age of 3.2 (1.0-6.7) years (n = 62) and serum liver biochemistry. Overall, NEC patients had less end-jejunostomies and autologous intestinal reconstruction surgeries performed compared to non-NEC patients (< 0.05), while remaining small bowel anatomy was comparable between groups. Cumulative weaning off PN was more frequent and duration of PN shorter among NEC patients (P < 0.05). Overall cumulative probability of histological IFALD was lower among NEC patients during whole follow-up period (P = 0.052) and at 10 years (P = 0.024). NEC patients had lower ALT and GGT levels at last follow-up (P < 0.05 for all). In univariate Cox regression analysis, absence of end-jejunostomy, NEC diagnosis, longer remaining small bowel length, multidisciplinary treatment and prematurity were predictive for weaning off PN, while NEC diagnosis and lower birth weight in addition to multidisciplinary care protected from histological IFALD. Neonates with NEC induced SBS reached enteral autonomy earlier than those with non-NEC SBS, which associated with more efficient resolution of histological IFALD among long-term survivors.
Collapse
Affiliation(s)
- Annika Mutanen
- grid.7737.40000 0004 0410 2071Department of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O Box 347, 00029 HUS Helsinki, Finland
| | - Ville Pöntinen
- grid.7737.40000 0004 0410 2071Department of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O Box 347, 00029 HUS Helsinki, Finland
| | - Riikka Gunnar
- grid.7737.40000 0004 0410 2071Department of Pediatric Gastroenterology, Pediatric Liver and Gut Research Group, Pediatric Research Center, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Laura Merras-Salmio
- grid.7737.40000 0004 0410 2071Department of Pediatric Gastroenterology, Pediatric Liver and Gut Research Group, Pediatric Research Center, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko P. Pakarinen
- grid.7737.40000 0004 0410 2071Department of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9, P.O Box 347, 00029 HUS Helsinki, Finland
| |
Collapse
|
40
|
Cui M, Liu W, Liu Q, Wang Y, Guo Z. Analysis of Cases of Neonatal Appendicitis from a Tertiary Care Unit. Indian J Pediatr 2022; 89:996-1002. [PMID: 35380382 DOI: 10.1007/s12098-022-04090-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/27/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To compare the clinical characteristics and prognosis of neonatal appendicitis (NA) alone and neonatal appendicitis with neonatal necrotizing enterocolitis (NEC) to improve the early recognition and diagnosis rate for this type of disease. METHODS Cases of appendicitis proved by operation and pathology younger than 28 d old in the authors hospital from 1990 to 2021 were retrospectively analyzed. According to whether combine with NEC, the cases were divided into two groups, analyzes the clinical characteristics. RESULTS A total of 48 patients were enrolled, 15 cases in the NA group and 33 cases in the NEC with NA group. The age of onset, time from onset of symptoms to surgical intervention were both earlier in the NEC with NA group (p < 0.05). The preoperative white blood cells (WBC) and platelets (PLT) in the NA group were higher (p < 0.05). NEC with NA was more likely to be complicated with neonatal pneumonia and sepsis before surgery (p < 0.05). The main clinical symptom of NA was abdominal distension, while the other was bloody stool. The positive rate of ultrasound was high before the operation. The perforation rate of the appendix was very high (NA 100% vs. NEC 57.6%). In the NA group, 100% underwent appendectomy and 78.8% in the NEC with NA group underwent terminal ileostomy and appendectomy. CONCLUSION The incidence of neonatal appendicitis is low. It is easily misdiagnosed as NEC. The perforation rate is very high, it is recommended to operate as soon as possible, and the prognosis is good.
Collapse
Affiliation(s)
- Mengying Cui
- Department of General Surgery & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Liangjiang Wing, Yubei District, Chongqing, 401122, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Liu
- Department of General Surgery & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Liangjiang Wing, Yubei District, Chongqing, 401122, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Qingshuang Liu
- Department of General Surgery & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Liangjiang Wing, Yubei District, Chongqing, 401122, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Wang
- Department of General Surgery & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Liangjiang Wing, Yubei District, Chongqing, 401122, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenhua Guo
- Department of General Surgery & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Liangjiang Wing, Yubei District, Chongqing, 401122, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
41
|
Gagné D, Shajari E, Thibault MP, Noël JF, Boisvert FM, Babakissa C, Levy E, Gagnon H, Brunet MA, Grynspan D, Ferretti E, Bertelle V, Beaulieu JF. Proteomics Profiling of Stool Samples from Preterm Neonates with SWATH/DIA Mass Spectrometry for Predicting Necrotizing Enterocolitis. Int J Mol Sci 2022; 23:ijms231911601. [PMID: 36232903 PMCID: PMC9569884 DOI: 10.3390/ijms231911601] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening condition for premature infants in neonatal intensive care units. Finding indicators that can predict NEC development before symptoms appear would provide more time to apply targeted interventions. In this study, stools from 132 very-low-birth-weight (VLBW) infants were collected daily in the context of a multi-center prospective study aimed at investigating the potential of fecal biomarkers for NEC prediction using proteomics technology. Eight of the VLBW infants received a stage-3 NEC diagnosis. Stools collected from the NEC infants up to 10 days before their diagnosis were available for seven of them. Their samples were matched with those from seven pairs of non-NEC controls. The samples were processed for liquid chromatography-tandem mass spectrometry analysis using SWATH/DIA acquisition and cross-compatible proteomic software to perform label-free quantification. ROC curve and principal component analyses were used to explore discriminating information and to evaluate candidate protein markers. A series of 36 proteins showed the most efficient capacity with a signature that predicted all seven NEC infants at least a week in advance. Overall, our study demonstrates that multiplexed proteomic signature detection constitutes a promising approach for the early detection of NEC development in premature infants.
Collapse
Affiliation(s)
- David Gagné
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Elmira Shajari
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Marie-Pier Thibault
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Noël
- PhenoSwitch Bioscience Inc., 975 Rue Léon-Trépanier, Sherbrooke, QC J1G 5J6, Canada
| | - François-Michel Boisvert
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Corentin Babakissa
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Emile Levy
- Research Center, Centre Hospitalier Universitaire Ste-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Hugo Gagnon
- PhenoSwitch Bioscience Inc., 975 Rue Léon-Trépanier, Sherbrooke, QC J1G 5J6, Canada
| | - Marie A. Brunet
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - David Grynspan
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Colombia, Vancouver, BC V6T 2B5, Canada
| | - Emanuela Ferretti
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Valérie Bertelle
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
42
|
The administration of a pre-digested fat-enriched formula prevents necrotising enterocolitis-induced lung injury in mice. Br J Nutr 2022; 128:1050-1063. [PMID: 34632971 PMCID: PMC8995403 DOI: 10.1017/s0007114521004104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Necrotising enterocolitis (NEC) is a devastating gastrointestinal disease of prematurity that typically develops after the administration of infant formula, suggesting a link between nutritional components and disease development. One of the most significant complications that develops in patients with NEC is severe lung injury. We have previously shown that the administration of a nutritional formula that is enriched in pre-digested Triacylglyceride that do not require lipase action can significantly reduce the severity of NEC in a mouse model. We now hypothesise that this 'pre-digested fat (PDF) system' may reduce NEC-associated lung injury. In support of this hypothesis, we now show that rearing newborn mice on a nutritional formula based on the 'PDF system' promotes lung development, as evidenced by increased tight junctions and surfactant protein expression. Mice that were administered this 'PDF system' were significantly less vulnerable to the development of NEC-induced lung inflammation, and the administration of the 'PDF system' conferred lung protection. In seeking to define the mechanisms involved, the administration of the 'PDF system' significantly enhanced lung maturation and reduced the production of reactive oxygen species (ROS). These findings suggest that the PDF system protects the development of NEC-induced lung injury through effects on lung maturation and reduced ROS in the lung and also increases lung maturation in non-NEC mice.
Collapse
|
43
|
Dias Vieira LG, da Silva VM, Lopes MVDO, Gomes de Souza NM, Gomes Guedes N, Torres França AC, Sales da Silva AC. Accuracy of clinical indicators of the nursing diagnosis of dysfunctional gastrointestinal motility in infants. J Child Health Care 2022; 26:343-354. [PMID: 33913358 DOI: 10.1177/13674935211014744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study aimed to analyze the accuracy of the clinical indicators of the nursing diagnosis of dysfunctional gastrointestinal motility in infants from neonatal units and identify their association with clinical variables. This is a study of the diagnostic accuracy of clinical indicators of the diagnosis of dysfunctional gastrointestinal motility, with a cross-sectional design, performed on 228 hospitalized infants in neonatal units. A high prevalence of dysfunctional gastrointestinal motility was identified in the studied population. Regarding accuracy measures, clinical indicators such as increased gastric residual, changes in bowel sounds, bile-colored gastric residual, regurgitation, absence of flatus, and hard and formed stool were useful to discriminate between infants with and without dysfunctional gastrointestinal motility. The findings can help nurses during the diagnostic process, as they identify which defining characteristics can be used to confirm or rule out the probability of occurrence of the diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ana Caroline Sales da Silva
- Neonatal Intensive Care in Fortaleza General Hospital, 28121Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
44
|
刘 欣, 刘 利, 蒋 海, 赵 常, 何 海. [Establishment of a nomogram model for predicting necrotizing enterocolitis in very preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:778-785. [PMID: 35894193 PMCID: PMC9336614 DOI: 10.7499/j.issn.1008-8830.2202093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/25/2022] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To investigate the risk factors for necrotizing enterocolitis (NEC) in very preterm infants and establish a nomogram model for predicting the risk of NEC. METHODS A total of 752 very preterm infants who were hospitalized from January 2015 to December 2021 were enrolled as subjects, among whom 654 were born in 2015-2020 (development set) and 98 were born in 2021 (validation set). According to the presence or absence of NEC, the development set was divided into two groups: NEC (n=77) and non-NEC (n=577). A multivariate logistic regression analysis was used to investigate the independent risk factors for NEC in very preterm infants. R software was used to plot the nomogram model. The nomogram model was then validated by the data of the validation set. The receiver operating characteristic (ROC) curve, the Hosmer-Lemeshow goodness-of-fit test, and the calibration curve were used to evaluate the performance of the nomogram model, and the clinical decision curve was used to assess the clinical practicability of the model. RESULTS The multivariate logistic regression analysis showed that neonatal asphyxia, sepsis, shock, hypoalbuminemia, severe anemia, and formula feeding were independent risk factors for NEC in very preterm infants (P<0.05). The ROC curve of the development set had an area under the curve (AUC) of 0.833 (95%CI: 0.715-0.952), and the ROC curve of the validation set had an AUC of 0.826 (95%CI: 0.797-0.862), suggesting that the nomogram model had a good discriminatory ability. The calibration curve analysis and the Hosmer-Lemeshow goodness-of-fit test showed good accuracy and consistency between the predicted value of the model and the actual value. CONCLUSIONS Neonatal asphyxia, sepsis, shock, hypoalbuminemia, severe anemia, and formula feeding are independent risk factors for NEC in very preterm infant. The nomogram model based on the multivariate logistic regression analysis provides a quantitative, simple, and intuitive tool for early assessment of the development of NEC in very preterm infants in clinical practice.
Collapse
MESH Headings
- Asphyxia/complications
- Child
- Enterocolitis, Necrotizing/complications
- Enterocolitis, Necrotizing/etiology
- Female
- Fetal Growth Retardation
- Humans
- Hypoalbuminemia
- Infant
- Infant, Newborn
- Infant, Newborn, Diseases
- Infant, Premature
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/etiology
- Nomograms
- Sepsis/complications
Collapse
|
45
|
Development of artificial neural networks for early prediction of intestinal perforation in preterm infants. Sci Rep 2022; 12:12112. [PMID: 35840701 PMCID: PMC9287325 DOI: 10.1038/s41598-022-16273-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/07/2022] [Indexed: 11/09/2022] Open
Abstract
Intestinal perforation (IP) in preterm infants is a life-threatening condition that may result in serious complications and increased mortality. Early Prediction of IP in infants is important, but challenging due to its multifactorial and complex nature of the disease. Thus, there are no reliable tools to predict IP in infants. In this study, we developed new machine learning (ML) models for predicting IP in very low birth weight (VLBW) infants and compared their performance to that of classic ML methods. We developed artificial neural networks (ANNs) using VLBW infant data from a nationwide cohort and prospective web-based registry. The new ANN models, which outperformed all other classic ML methods, showed an area under the receiver operating characteristic curve (AUROC) of 0.8832 for predicting IP associated with necrotizing enterocolitis (NEC-IP) and 0.8797 for spontaneous IP (SIP). We tested these algorithms using patient data from our institution, which were not included in the training dataset, and obtained an AUROC of 1.0000 for NEC-IP and 0.9364 for SIP. NEC-IP and SIP in VLBW infants can be predicted at an excellent performance level with these newly developed ML models. https://github.com/kdhRick2222/Early-Prediction-of-Intestinal-Perforation-in-Preterm-Infants.
Collapse
|
46
|
Fetal Doppler Evaluation to Predict NEC Development. J Pers Med 2022; 12:jpm12071042. [PMID: 35887539 PMCID: PMC9323983 DOI: 10.3390/jpm12071042] [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: 06/07/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Antenatal factors play a role in NEC pathogenesis. This study aimed to investigate the predictive value of fetal ductus venosus doppler (DV) for NEC in fetal growth restriction fetuses (FGRF) and to assess the predictive accuracy of IG21 and Fenton curves in NEC development. Data from FGRF, postnatal findings, and Doppler characteristics were collected between 2010 and 2020 at a single center. Patients were then divided into two groups (i.e., with and without NEC). Bivariate and multivariate analyses were performed. We identified 24 cases and 30 controls. Absent or reversed end-diastolic flow (AREDF) and increased resistance in the DV were more impaired in cases (p < 0.05). Although the median birthweight was not different, the Fenton z-score was lower in NEC (p < 0.05). Fetal cardiopulmonary resuscitation, synchronized intermittent mandatory ventilation, neonatal respiratory distress, persistent patent ductus arteriosus (PDA), and inotropic support were more frequent in the NEC group. Furthermore, NEC patients had lower white blood cells (WBC) (p < 0.05). The predictive model for NEC (model 4), including Fenton z-score, WBC, PDA, and DV had an AUC of 84%. Fetal Doppler findings proved effective in predicting NEC in FGR. The Fenton z-score was the most predictive factor considering the fetal growth assessment showing high sensitivity.
Collapse
|
47
|
Bao ZK, Mi YH, Xiong XY, Wang XH. Sulforaphane Ameliorates the Intestinal Injury in Necrotizing Enterocolitis by Regulating the PI3K/Akt/GSK-3 β Signaling Pathway. Can J Gastroenterol Hepatol 2022; 2022:6529842. [PMID: 35600210 PMCID: PMC9117068 DOI: 10.1155/2022/6529842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Necrotizing enterocolitis (NEC) is a serious neonatal disease; this study aims to investigate the role of sulforaphane (SFN) in NEC-induced intestinal injury. Methods An animal model of NEC was established in newborn mice and intragastrically administrated with SFN; then, the general status and survival of the mice were observed. H&E staining was used to observe the pathological changes of intestinal tissues. ELISA, immunohistochemical staining, and flow cytometry assays were used to detect the levels of inflammatory factors, including TNF-α, IL-6, and IL-17, the expression of Bax, Bcl-2, TLR4, and NF-κB, and the percentages of the Th17 and Treg cells, respectively. GSK-3β expression levels were measured by immunofluorescence. IEC-6 and FHC cells were induced with LPS to mimic NEC in vitro and coincubated with SFN; then, the inflammatory factor levels and cell apoptosis rate were detected. Finally, Western blot was used to assess the expression of PI3K/Akt/GSK-3β pathway-related proteins in vitro and in vivo. Results SFN improved the survival rate of NEC mice during modeling, alleviated the severity of the intestinal injury, and reduced the proportion of Th17/Treg cells. SFN could inhibit TLR4 and NF-κB levels, decrease the release of inflammatory factors TNF-α and IL-6, suppress Bax expression, increase Bcl-2 expression, and inhibit apoptosis both in in vitro and in vivo models of NEC. Meanwhile, SFN regulated the expression of PI3K/Akt/GSK-3β pathway-related proteins in vitro and in vivo. Conclusion SFN relieved the inflammatory response and apoptosis by regulating the PI3K/Akt/GSK-3β signaling pathway, thereby alleviating NEC in model mice and cells.
Collapse
Affiliation(s)
- Zhong-Kun Bao
- Department of Radiology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Hong Mi
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, China
| | - Xiao-Yu Xiong
- Department of Neonatology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xin-Hong Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Sun X, Xu L, Ma S, Chen L, Tang R, Li D, Hu F, Wang T, Gong Y, Zhou H, Wang J. Reduced Incidence of Necrotizing Enterocolitis due to the Anti-Inflammatory Effects of CXCL14 in Intestinal Tissue. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:1322172. [PMID: 35463668 PMCID: PMC9023168 DOI: 10.1155/2022/1322172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/06/2022] [Accepted: 03/12/2022] [Indexed: 11/18/2022]
Abstract
Bioinformatic analysis indicated that downregulated CXCL14 will occur in the intestinal tissue of patients with necrotizing enterocolitis (NEC). To reveal the relationship between CXCL14 and mucosal immune regulation, we designed and implemented the experiments to explore the potential function of CXCL14 in the pathogenesis of NEC. Firstly, this study confirmed that the expression of CXCL14 decreased in the intestinal tract of NEC children. Secondly, the experiments results showed that CXCL14 could ameliorate the inflammatory injury of intestinal tissue through the suppressive effect on the expression of TNF-α and INF-γ in vivo. Finally, we explained that activation of the TLR4 can reduce the expression level of CXCL14 in the intestinal tissue of mouse pups. Collectively, our study suggested that CXCL14 may negatively regulate the inflammatory response in intestinal tissue and play an essential role in NEC development and progression.
Collapse
Affiliation(s)
- Xu Sun
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Lingqi Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Shurong Ma
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Lulu Chen
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Ruze Tang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Dashuang Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Fangjie Hu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Ting Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Yuan Gong
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Huiting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| |
Collapse
|
49
|
Hwang M, Tierradentro-García LO, Dennis RA, Anupindi SA. The role of ultrasound in necrotizing enterocolitis. Pediatr Radiol 2022; 52:702-715. [PMID: 34654968 DOI: 10.1007/s00247-021-05187-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/07/2021] [Accepted: 08/12/2021] [Indexed: 01/10/2023]
Abstract
Ultrasound has proved to be a useful modality for enhancing the diagnostic accuracy of necrotizing enterocolitis and associated complications. The standard imaging algorithm for evaluating necrotizing enterocolitis includes radiographs and clinical symptoms, the combination of which constitutes the Bell criteria. Major limitations of using the Bell criteria for diagnosing and clinically managing necrotizing enterocolitis include low diagnostic accuracy of radiographs and nonspecific symptomatology of preterm infants. In this regard, US can offer additional insights into bowel health by helping to characterize bowel motility, echogenicity, thickness, pneumatosis and perfusion. Extramural findings such as portal venous gas, nature and extent of ascites, and pneumoperitoneum can also be assessed. Recently, contrast-enhanced US was explored in a case series of preterm bowel disease and its diagnostic utility warrants further investigation. This article reviews the US features of necrotizing enterocolitis and highlights the role of US as a complement to radiographs, as well as the emerging use of contrast-enhanced US in necrotizing enterocolitis.
Collapse
Affiliation(s)
- Misun Hwang
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA. .,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Luis O Tierradentro-García
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Rebecca A Dennis
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha A Anupindi
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Abramov A, Luks VL, De Bie F, Hwang R, Allukian M, Nace GW. Pneumatosis intestinalis in children beyond the neonatal period: is it always benign? Pediatr Surg Int 2022; 38:399-407. [PMID: 34837497 DOI: 10.1007/s00383-021-05048-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE The significance and management of pediatric pneumatosis intestinalis (PI) remains poorly defined. We sought to add clarity in children beyond the neonatal period. METHODS Pediatric patients 3 months-18 years admitted to a quaternary children's hospital with a diagnosis of PI were included in this retrospective study. Pathologic PI was defined as irreversible, transmural intestinal ischemia. RESULTS 167 children were identified with PI. Of these children, 155 (92.8%) had benign PI and 12 (7.2%) developed pathologic PI. The most common underlying diagnosis for pathologic PI was global developmental delay (75%), although we identified a spectrum of underlying diagnoses at risk for PI. Physical exam notable for abdominal distension (p = 0.023) or guarding (p = 0.028), and imaging with portal venous gas (p < 0.001) or bowel distension (p = 0.001) were significantly associated with pathologic PI. Only 6.6% of all children underwent an operation. For those undergoing non-surgical management of benign PI, 75% of children received antibiotics and average duration of bowel rest was 6.8 days. CONCLUSIONS PI in children is primarily a benign phenomenon and often does not warrant surgical intervention. Bowel rest and antibiotics are therapeutic strategies frequently used in the treatment of this finding.
Collapse
Affiliation(s)
- Alexey Abramov
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Valerie L Luks
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Felix De Bie
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Rosa Hwang
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Myron Allukian
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Gary W Nace
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|