1
|
Chen Y, Chen W, Dai Y, Yan X, Jiang C, Zhang F, Zhang M, Hu X, Zhao J, Wu T, Li S, Han S, Chen X. Human breast milk-derived phospholipid DOPE ameliorates intestinal injury associated with NEC by inhibiting ferroptosis. Food Funct 2024; 15:10811-10822. [PMID: 39403969 DOI: 10.1039/d4fo03904a] [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: 10/29/2024]
Abstract
Neonatal necrotizing enterocolitis (NEC) is a severe inflammatory bowel disease that commonly affects premature infants. Breastfeeding has been proven to be one of the most effective methods for preventing NEC. However, the specific role of lipids, the second major nutrient category in human breast milk (HBM), in intestinal development remains unclear. Our preliminary lipidomic analysis of the HBM lipidome revealed that dioleoyl phosphatidylethanolamine (DOPE) is not only abundant but also shows high solubility in lipids, endowing it with significant biological utility. Experimental results confirmed that DOPE significantly reduces the mortality of neonatal rats, ameliorates impairment of intestinal barrier function, and alleviates the expression of intestinal inflammatory factors IL-1β and IL-6. Furthermore, DOPE promotes the migration and proliferation of intestinal epithelial cells, thereby enhancing the integrity of the intestinal barrier function in vitro. The progression of NEC is linked with the onset of ferroptosis. Our cellular-level analysis of lipid peroxide and iron ion concentrations revealed that DOPE significantly reduces the indicators of ferroptosis, while also modulating the expression of pivotal ferroptosis-associated factors, including SLC7A11, GPX4, and ACSL4. Hence, this research on DOPE is expected to provide novel insights into the bioactive lipids present in HBM.
Collapse
Affiliation(s)
- Yanjie Chen
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Wenjuan Chen
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Yu Dai
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Xiangyun Yan
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Chengyao Jiang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Fan Zhang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Min Zhang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Xiaoshan Hu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Juyi Zhao
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Tingyue Wu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Shushu Li
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Shuping Han
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| | - Xiaohui Chen
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Gibbons JA, Worthington LAM, Chiu EG, Kates HR, Carter RR, Nelson R, Zhang M, Garrett TJ, Ho TTB. Severe anemia in preterm infants associated with increased bacterial virulence potential and metabolic disequilibrium. Pediatr Res 2024:10.1038/s41390-024-03669-4. [PMID: 39438713 DOI: 10.1038/s41390-024-03669-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/16/2024] [Accepted: 09/15/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Anemia in preterm infants is associated with gut dysbiosis and necrotizing enterocolitis. Our study aimed to identify the bacterial functions and metabolites that can explain the underlying mechanisms of anemia associated disease conditions. METHODS We conducted a case control study in preterm infants with cases having a hematocrit ≤ 25%. The control infants were matched by birth gestational age and weight. Fecal samples were collected before, at the onset, and after the onset of anemia in cases and with matched postnatal age in controls for metagenomics and metabolomics analyzes. RESULTS 18 anemic and 20 control infants with fecal samples collected at 17 days, 5 weeks, and 7 weeks postnatal age were included. Virulence factor potential, decrease in beta diversity evolution, and larger changes in metabolome were associated with severe anemia. Metabolite abundances of N-acetylneuraminate and butyrobetaine were associated with virulence factor potential. Anemic group had decreased prostaglandin and lactic acid levels. CONCLUSION Fecal omics data showed that severe anemia is associated with a pro-inflammatory gut microbiota with more virulent and less commensal anaerobic bacterial activities. Future studies can examine the link between anemia-associated dysbiosis and clinical outcomes and predict an infant-specific hematocrit threshold that negatively affects clinical outcomes. IMPACT Severe anemia in preterm infants contributes to a pro-inflammatory gut with greater bacterial virulence and less commensal bacterial activities. The multiomics approach using non-invasive fecal biospecimens identified functional and metabolic changes in the gut microbiota and these mechanistic changes are plausible explanations for anemia-associated disease conditions in preterm infants. Our findings identified biological changes of the gut environment in severely anemic preterm infants that can offer guidance for clinical management.
Collapse
Affiliation(s)
- Justin A Gibbons
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Leigh-Anne M Worthington
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Elizabeth G Chiu
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Heather R Kates
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Rico R Carter
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Rachel Nelson
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Min Zhang
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Thao T B Ho
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
3
|
Çıplak G, Sarı FN, Erten EE, Azılı MN, Bostancı SA, Tayman C, Alyamaç Dizdar E, Şenel E. Does serum albumin at the onset of necrotisıng enterocolitis predict severe disease in preterm infants? Pediatr Surg Int 2024; 40:267. [PMID: 39382780 DOI: 10.1007/s00383-024-05850-6] [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: 09/30/2024] [Indexed: 10/10/2024]
Abstract
OBJECTIVE To investigate whether laboratory markers obtained at the onset of necrotising enterocolitis (NEC) predict the severity of the disease in preterm infants. METHODS Prospective cohort study conducted in a tertiary referance hospital. A total of 88 preterm infants were included in the study. Of those, 60 infants had the diagnosis of severe NEC, while the remaining 28 infants constituted the non-severe NEC group. Severe NEC was defined as surgical NEC or NEC-related mortality. Infants with and without severe NEC were compared in terms of demographic, clinical and laboratory characteristics. RESULTS At the onset of disease, infants with severe NEC noted to have lower platelet count and serum ALB levels (p = 0.011, p = 0.004; respectively), whereas higher CRP, and serum lactate levels (p = 0.009, p = 0.008; respectively). Multiple binary logistic regression analyses showed that CRP (1.03(1.01-1.05), p = 0.024) and serum albumin level (0.16(0.04-0.64), p = 0.010) were statistically significant independent risk factors for severe NEC. The optimal cut-off value for the serum ALB level was found to be 23 g/L with 52% sensitivity (95%CI: 37-68%) and 84% specificity (95%CI: 60-97%) (AUC 0.727; p = 0.002). CONCLUSION Serum ALB level at NEC onset might be a reliable biomarker for severe disease in preterm infants.
Collapse
Affiliation(s)
- Gökçe Çıplak
- Department of Neonatology, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey.
| | - Fatma Nur Sarı
- Department of Neonatology, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Elif Emel Erten
- Department of Pediatric Surgery, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Müjdem Nur Azılı
- Faculty of Medicine, Department of Pediatric Surgery, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Süleyman Arif Bostancı
- Department of Pediatric Surgery, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Cüneyt Tayman
- Department of Neonatology, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Evrim Alyamaç Dizdar
- Department of Neonatology, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Emrah Şenel
- Faculty of Medicine, Department of Pediatric Surgery, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
4
|
Sha C, Jin Z, Ku SY, Kogosov AS, Yu S, Bergese SD, Hsieh H. Necrotizing Enterocolitis and Neurodevelopmental Impairments: Microbiome, Gut, and Brain Entanglements. Biomolecules 2024; 14:1254. [PMID: 39456187 PMCID: PMC11505939 DOI: 10.3390/biom14101254] [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: 08/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
There is significant communication and interdependence among the gut, the microbiome, and the brain during development. Diseases, such as necrotizing enterocolitis (NEC), highlight how injury to the immature gastrointestinal tract leads to long-term neurological consequences, due to vulnerabilities of the brain in the early stages of life. A better understanding of the developing gut-microbiota-brain axis is needed to both prevent and treat the devastating consequences of these disease processes. The gut-microbiota-brain axis is a bidirectional communication pathway that includes metabolic, nervous, endocrine, and immune components. In this review, we discuss gut development, microbiome colonization and maturation, and the interactions that influence neurodevelopment in the context of NEC. We describe the components of the gut-brain axis and how the microbiome is an integral member of this relationship. Finally, we explore how derangements within the microbiome and gut-microbiota-brain axis affect the normal development and function of the other systems and long-term neurodevelopmental consequences for patients.
Collapse
Affiliation(s)
- Cuilee Sha
- Department of Pharmacological Sciences, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA;
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Stella Y. Ku
- Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Ann S. Kogosov
- Renaissance School of Medicine, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sun Yu
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sergio D. Bergese
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Helen Hsieh
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| |
Collapse
|
5
|
Weller JH, Scheese D, Tragesser C, Yi PH, Alaish SM, Hackam DJ. Artificial Intelligence vs. Doctors: Diagnosing Necrotizing Enterocolitis on Abdominal Radiographs. J Pediatr Surg 2024; 59:161592. [PMID: 38955625 PMCID: PMC11401766 DOI: 10.1016/j.jpedsurg.2024.06.001] [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: 04/09/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Radiographic diagnosis of necrotizing enterocolitis (NEC) is challenging. Deep learning models may improve accuracy by recognizing subtle imaging patterns. We hypothesized it would perform with comparable accuracy to that of senior surgical residents. METHODS This cohort study compiled 494 anteroposterior neonatal abdominal radiographs (214 images NEC, 280 other) and randomly divided them into training, validation, and test sets. Transfer learning was utilized to fine-tune a ResNet-50 deep convolutional neural network (DCNN) pre-trained on ImageNet. Gradient-weighted Class Activation Mapping (Grad-CAM) heatmaps visualized image regions of greatest relevance to the pretrained neural network. Senior surgery residents at a single institution examined the test set. Resident and DCNN ability to identify pneumatosis on radiographic images were measured via area under the receiver operating curves (AUROC) and compared using DeLong's method. RESULTS The pretrained neural network achieved AUROC of 0.918 (95% CI, 0.837-0.978) with an accuracy of 87.8% with five false negative and one false positive prediction. Heatmaps confirmed appropriate image region emphasis by the pretrained neural network. Senior surgical residents had a median area under the receiver operating curve of 0.896, ranging from 0.778 (95% CI 0.615-0.941) to 0.991 (95% CI 0.971-0.999) with zero to five false negatives and one to eleven false positive predictions. The deep convolutional neural network performed comparably to each surgical resident's performance (p > 0.05 for all comparisons). CONCLUSIONS A deep convolutional neural network trained to recognize pneumatosis can quickly and accurately assist clinicians in promptly identifying NEC in clinical practice. LEVEL OF EVIDENCE III (study type: Study of Diagnostic Test, study of nonconsecutive patients without a universally applied "gold standard").
Collapse
Affiliation(s)
- Jennine H Weller
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Scheese
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cody Tragesser
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul H Yi
- Malone Center for Engineering in Healthcare, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Samuel M Alaish
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
Hu J, Yao Q, Zhao L. Evidences and perspectives on the association between gut microbiota and sepsis: A bibliometric analysis from 2003 to 2023. Heliyon 2024; 10:e37921. [PMID: 39315201 PMCID: PMC11417584 DOI: 10.1016/j.heliyon.2024.e37921] [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: 05/18/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Background In the last two decades, the role of the gut microbiome in the development, maintenance, and outcome of sepsis has received increased attention; however, few descriptive studies exist on its research focus, priorities, and future prospects. This study aimed to identify the current state, evolution, and emerging trends in the field of gut microbiota and sepsis using bibliometric analysis. Methods All publications on sepsis and gut microbiota were retrieved from the Web of Science Core Collection and included in this study. VOSviewer, CiteSpace, and the Web of Science online analysis platform were used to visualize trends based on publication country, institution, author, journal, and keywords. Results A total of 1,882 articles on sepsis-related gut microbiota were screened, mainly from 95 countries or regions and 2,581 institutions. The United States and China contributed the most to this research field, with 521 (27.683 %) and 376 (19.979 %) articles, respectively. Scientists from the University of California were the most prolific, publishing 63 (3.348 %) articles. Cani PD published papers with the highest H-index, establishing himself as a leader in the field. The most publications were published in the journals "Nutrients" and "PLOS One." The journals with the most co-citations were "PLOS One," "Nature," and "Gut." The most used keywords were prebiotics, gut microbiota, and sepsis. The keyword burst research analysis revealed that research on treatment strategies based on the intestinal microbiota, intestine-liver axis, and regulatory mechanisms of bacterial metabolites are currently hot directions. Conclusion This study presents a global overview of the current state and potential trends in the field of sepsis-related gut microbiota. This study identified hot research sub-directions and new trends through comparison and analysis, which will aid in the development of this field.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou City, 310003, China
| | - Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Linjun Zhao
- Department of Emergency, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Rd, Hangzhou City, 310006, China
| |
Collapse
|
7
|
Ma Y, Zhang Y, Liu X, Yang X, Guo H, Ding X, Ye C, Guo C. Deletion of CD38 mitigates the severity of NEC in experimental settings by modulating macrophage-mediated inflammation. Redox Biol 2024; 77:103336. [PMID: 39366069 PMCID: PMC11489332 DOI: 10.1016/j.redox.2024.103336] [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: 07/27/2024] [Accepted: 08/29/2024] [Indexed: 10/06/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a form of potentially lethal gastrointestinal inflammation that primarily affects preterm neonates. It is crucial to recognize that, while the disease carries significant risks, timely and effective medical intervention can greatly enhance the chances of survival. Additionally, NEC is closely linked to the activation of macrophages, highlighting the complex interplay between the immune response and disease progression. CD38, acting as an ectoenzyme, catalyzes the hydrolysis of NAD+ to produce cyclic ADP-ribose (cADPR), a reaction critical for modulating cellular redox balance and energy homeostasis. This enzymatic activity is particularly pertinent in the context of necrotizing enterocolitis (NEC). In this research, we investigated whether CD38 deletion can elevate NAD+ levels to reduce macrophage-mediated inflammation and improve NEC severity. We show that NEC patients was associated with the increased CD38 expression in intestine and blood. These results were also observed in NEC mice, and CD38 deletion ameliorated NEC intestinal injury. Mechanistically, CD38 deletion elevated NAD+ levels that reduced oxidative stress and intestinal inflammation. Furthermore, CD38 deletion promoted M2 macrophage polarization, inhibited macrophage activation and phagocytosis ability. Thus, our results reveal a critical role for CD38 as an intracellular immune regulator for regulating macrophage activation and intestinal inflammation in NEC. Targeting CD38 and NAD+ signal maybe a promising strategy for treatment of NEC.
Collapse
Affiliation(s)
- Yue Ma
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China
| | - Yunfei Zhang
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xinli Liu
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xinyi Yang
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; Department of Animal Center, Chongqing Medical University, Chongqing, China
| | - Hongjie Guo
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xionghui Ding
- Department of Animal Center, Chongqing Medical University, Chongqing, China; Department of General Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Cuilian Ye
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Chunbao Guo
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China.
| |
Collapse
|
8
|
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
|
9
|
Wu S, Ren X, Zhuang Y, Shen C, Zhu H, Cao Y, Zhang X, Chen G. Peripheral Lymphocyte Changes Associate With the Progression of Necrotizing Enterocolitis in Infants. J Surg Res 2024; 301:215-223. [PMID: 38959630 DOI: 10.1016/j.jss.2024.04.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Immune factors are important antecedents in the pathophysiology of necrotizing enterocolitis (NEC). However, studies on the peripheral blood lymphocyte subsets changes in NEC patients among different Bell stages and in patients requiring surgery are scarce. METHODS 34 infants with NEC and 33 age-matched controls were included. Peripheral blood was collected within 48 h after NEC diagnosis. Peripheral blood B and T lymphocytes subsets were detected by 12-color flow cytometry. Cell ratios were calculated, and their relationship to disease severity and their roles as indicators for surgery were assessed. RESULTS NEC patients showed elevated percentages of unSwB cells (CD27+IgD+ unswitched memory/activated B cells)/B cells, SwB cells (CD27+IgD-switched memory B cells)/B cells, CD8+ T (CD3+CD8+ T cells)/T cells, Tem (CD45RA-CCR7-effector memory T cells)/CD4+ T cells, Tem/CD8+ T cells and decreased Bn (CD27-IgD+ naïve B cells)/B cells, CD4+T (CD3+CD4+ T cells)/T cells, CD45RA+ CCR7+ naïve T cells (CD45RA+CCR7+ naïve T cells)/CD8+T cells. Compared to NEC patients at BELL stage I + II, patients at BELL stage III showed increased percentages of SwB cells/B cells, antibody secreting cell (ASC, CD3-CD20-CD27high CD38high ASCs)/B cells and Tem/CD4+ T cells, and decreased percentages of CD45RA+CCR7+ naïve T cells/CD4+ T cells. The Receiver Operating Characteristic Curve analysis showed that the sensitivity of ASC/B cells ratio (0.52%) is 86.67% and the specificity of Tem/CD4+T ratio (5.22%) is 100%, indicating that NEC patients required surgery. CONCLUSIONS The severity of NEC exhibits codirectional changes with the maturation of B and T lymphocytes, especially CD4+ T cells. The increased ASC/B and Tem/CD4+ T cells could serve as potential indicators for NEC patients requiring surgery.
Collapse
Affiliation(s)
- Shaojing Wu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Xue Ren
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yuxiu Zhuang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Chun Shen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Haitao Zhu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yun Cao
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Xiaoming Zhang
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Gong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China.
| |
Collapse
|
10
|
Ding XP, Hu XW, Chen S, Guo L, Wang ZL, He Y, Li LQ, Tang WY. Risk factors for necrotizing enterocolitis in small-for-gestational-age infants: a matched case-control study. Sci Rep 2024; 14:19098. [PMID: 39154081 PMCID: PMC11330453 DOI: 10.1038/s41598-024-70351-4] [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: 11/21/2023] [Accepted: 08/14/2024] [Indexed: 08/19/2024] Open
Abstract
Few studies have focused on the risk factors for necrotizing enterocolitis (NEC) in small for gestational age (SGA) infants. The aim of this study was to identify the risk factors for NEC in SGA newborns. This study included consecutive SGA neonates admitted to a tertiary hospital in Jiangxi Province, China from Jan 2008 to Dec 2022. Patients with NEC (Bell's stage ≥ II) were assigned to the NEC group. Gestational age- and birth weight-matched non-NEC infants born during the same period at the same hospital were assigned to the control group. The risk factors associated with NEC were analyzed with univariate and logistic regression models. During the study period, 2,912 SGA infants were enrolled, 150 (5.15%) of whom developed NEC. In total, 143 patients and 143 controls were included in the NEC and control groups, respectively. Logistic regression analysis revealed that sepsis (OR 2.399, 95% CI 1.271-4.527, P = 0.007) and anemia (OR 2.214, 95% CI 1.166-4.204, P = 0.015) might increase the incidence of NEC in SGA infants and that prophylactic administration of probiotics (OR 0.492, 95% CI 0.303-0.799, P = 0.004) was a protective factor against NEC. Therefore, sepsis, anemia and a lack of probiotic use are independent risk factors for NEC in SGA infants.
Collapse
Affiliation(s)
- Xiang-Ping Ding
- Department of Neonatology, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330103, People's Republic of China
| | - Xiang-Wen Hu
- Department of Neonatology, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330103, People's Republic of China
| | - Shi Chen
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Lu Guo
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Zheng-Li Wang
- Department of Neonatology, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330103, People's Republic of China
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Yu He
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Lu-Quan Li
- Department of Neonatology, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330103, People's Republic of China
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Wen-Yan Tang
- Department of Neonatology, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330103, People's Republic of China.
- Jiangxi Maternal and Child Health Hospital, Maternal and Child Health Hospital of Nanchang Medical College, Nan Chan, 330038, Jiangxi, 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
|
Tian B, Xu X, Li L, Tian Y, Liu Y, Mu Y, Lu J, Song K, Lv J, He Q, Zhong W, Xia H, Lan C. Epigenetic Insights Into Necrotizing Enterocolitis: Unraveling Methylation-Regulated Biomarkers. Inflammation 2024:10.1007/s10753-024-02054-x. [PMID: 38814387 DOI: 10.1007/s10753-024-02054-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/31/2024]
Abstract
Necrotizing enterocolitis (NEC) is a multifactorial gastrointestinal disease with high morbidity and mortality among premature infants. This study aimed to identify novel methylation-regulated biomarkers in NEC intestinal tissue through multiomics analysis. We analyzed DNA methylation and transcriptome datasets from ileum and colon tissues of patients with NEC. We identify methylation-related differential genes (MrDEGs) based on the rule that the degree of methylation in the promoter region is inversely proportional to RNA transcription. These MrDEGs included ADAP1, GUCA2A, BCL2L14, FUT3, MISP, USH1C, ITGA3, UNC93A and IL22RA1. Single-cell data revealed that MrDEGs were mainly located in the intestinal epithelial part of intestinal tissue. These MrDEGs were verified through Target gene bisulfite sequencing and RT-qPCR. We successfully identified and verified the ADAP1, GUCA2A, IL22RA1 and MISP, primarily expressed in intestinal epithelial villus cells through single-cell data. Through single-gene gene set enrichment analysis, we found that these genes participate mainly in the pathological process of T-cell differentiation and the suppression of intestinal inflammation in NEC. This study enhances our understanding of the pathogenesis of NEC and may promote the development of new precision medicine methods for NEC prediction and diagnosis.
Collapse
Affiliation(s)
- Bowen Tian
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaogang Xu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Lin Li
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Yan Tian
- Department of Anesthesiology, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi, China
| | - Yanqing Liu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Yide Mu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Jieting Lu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Kai Song
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Junjian Lv
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Qiuming He
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Wei Zhong
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China.
| | - Huimin Xia
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China.
| | - Chaoting Lan
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Whiteside SA, Odom John AR. Oops, the microbes did it again: Gut dysbiosis precedes late-onset meningitis. J Infect Dis 2024:jiae266. [PMID: 38779890 DOI: 10.1093/infdis/jiae266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Samantha A Whiteside
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Audrey R Odom John
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Elsayed Ramadan OM, Alruwaili MM, Alruwaili AN, Elsharkawy NB, Abdelaziz EM, Zaky ME, Shaban MM, Shaban M. Nursing practice of routine gastric aspiration in preterm infants and its link to necrotizing enterocolitis: is the practice still clinically relevant? BMC Nurs 2024; 23:333. [PMID: 38760751 PMCID: PMC11100149 DOI: 10.1186/s12912-024-01994-x] [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: 01/28/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
The practice of routine gastric residual aspiration in preterm infants remains controversial, with conflicting evidence regarding its impact on necrotizing enterocolitis (NEC). As front-line caregivers, nurses play a vital role in gastric aspiration procedures and must be informed by evidence. This quasi-experimental nursing study aimed to assess whether gastric aspiration is clinically relevant in reducing the risk of NEC in preterm infants.A total of 250 preterm infants from two NICUs in Egypt were allocated to the gastric aspiration (n = 125) and non-aspiration (n = 125) groups. Feeding practices, gastric residuals, and incidence/severity of NEC were compared between groups according to modified Bell's criteria. Risk factors were analyzed using multivariate regression. There were no significant baseline differences between the groups. The gastric residual attributes and feeding outcomes did not differ substantially from aspiration. The overall incidence of NEC was 14-15%, with no significant differences in the odds of onset or progression of NEC by stage between the groups. Lower gestational age and birth weight emerged as stronger predictors of NEC. Routine gastric aspiration does not appear to directly prevent or reduce the severity of NEC in this population. Although gastric residuals retain clinical importance, study findings question assumptions that aspiration protects against NEC and informs nursing practice. Evidence-based feeding protocols must continually evolve through ongoing research on modifiable risk factors for this devastating intestinal disease in preterm infants.
Collapse
Affiliation(s)
| | | | | | - Nadia Bassuoni Elsharkawy
- College of Nursing, Jouf University, Sakaka, Saudi Arabia, 72388
- Maternal and Newborn Health Nursing Department, Faculty of Nursing, Cairo University, Cairo, Egypt
| | - Enas Mahrous Abdelaziz
- College of Nursing, Jouf University, Sakaka, Saudi Arabia, 72388
- Psychiatric Mental Health Nursing Department, Faculty of Nursing, Cairo University, Cairo, Egypt
| | - Mohammed Elsayed Zaky
- College of Nursing, Jouf University, Sakaka, Saudi Arabia, 72388
- Medical Surgical Nursing Department, Faculty of Nursing, Cairo University, Cairo, Egypt
| | - Marwa Mamdouh Shaban
- Lecturer of Community Health Nursing, Faculty of Nursing, Cairo University, Cairo, Egypt
| | - Mostafa Shaban
- College of Nursing, Jouf University, Sakaka, Saudi Arabia, 72388
- Geriatric Nursing Department, Faculty of Nursing, Cairo University, Cairo, Egypt
| |
Collapse
|
15
|
古丽佐合热· 阿, 努尔亚· 热. [Application of near-infrared spectroscopy in the early identification of neonatal gastrointestinal diseases]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:523-528. [PMID: 38802915 PMCID: PMC11135058 DOI: 10.7499/j.issn.1008-8830.2311148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/19/2024] [Indexed: 05/29/2024]
Abstract
Necrotizing enterocolitis (NEC) is one of the most common gastrointestinal diseases in neonatal intensive care units, characterized by rapid progression and a high mortality rate. Local intestinal ischemia and hypoxia are significant contributors to NEC. Feeding intolerance (FI), which refers to a range of gastrointestinal manifestations arising from the inability to tolerate enteral nutrition, is one of the most common clinical issues in neonates, and preventing and treating FI is crucial for improving neonatal survival rates. Near-infrared spectroscopy is a clinical tool that can be used at the bedside to monitor regional oxygen saturation. It is non-invasive, reliable, and sustainable, and its feasibility and safety in assessing intestinal blood circulation have been validated. Early identification of intestinal ischemia and differentiation of FI from precursor symptoms of NEC, as well as predicting the occurrence of NEC, are extremely important for reducing intestinal injury and adverse long-term outcomes. In recent years, there has been new research progress related to the monitoring of intestinal tissue oxygen saturation and cerebral oxygen saturation for the early identification of FI and precursor symptoms of NEC, and this article provides a review of these developments.
Collapse
|
16
|
Harrison SP, Baumgarten SF, Chollet ME, Stavik B, Bhattacharya A, Almaas R, Sullivan GJ. Parenteral nutrition emulsion inhibits CYP3A4 in an iPSC derived liver organoids testing platform. J Pediatr Gastroenterol Nutr 2024; 78:1047-1058. [PMID: 38529852 DOI: 10.1002/jpn3.12195] [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: 11/15/2022] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVES Parenteral nutrition (PN) is used for patients of varying ages with intestinal failure to supplement calories. Premature newborns with low birth weight are at a high risk for developing PN associated liver disease (PNALD) including steatosis, cholestasis, and gallbladder sludge/stones. To optimize nutrition regimens, models are required to predict PNALD. METHODS We have exploited induced pluripotent stem cell derived liver organoids to provide a testing platform for PNALD. Liver organoids mimic the developing liver and contain the different hepatic cell types. The organoids have an early postnatal maturity making them a suitable model for premature newborns. To mimic PN treatment we used medium supplemented with either clinoleic (80% olive oil/20% soybean oil) or intralipid (100% soybean oil) for 7 days. RESULTS Homogenous HNF4a staining was found in all organoids and PN treatments caused accumulation of lipids in hepatocytes. Organoids exhibited a dose dependent decrease in CYP3A4 activity and expression of hepatocyte functional genes. The lipid emulsions did not affect overall organoid viability and glucose levels had no contributory effect to the observed results. CONCLUSIONS Liver organoids could be utilized as a potential screening platform for the development of new, less hepatotoxic PN solutions. Both lipid treatments caused hepatic lipid accumulation, a significant decrease in CYP3A4 activity and a decrease in the RNA levels of both CYP3A4 and CYP1A2 in a dose dependent manner. The presence of high glucose had no additive effect, while Clinoleic at high dose, caused significant upregulation of interleukin 6 and TLR4 expression.
Collapse
Affiliation(s)
- Sean P Harrison
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Saphira F Baumgarten
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub-Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Maria E Chollet
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Benedicte Stavik
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Anindita Bhattacharya
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Runar Almaas
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Demicheva E, Dordiuk V, Polanco Espino F, Ushenin K, Aboushanab S, Shevyrin V, Buhler A, Mukhlynina E, Solovyova O, Danilova I, Kovaleva E. Advances in Mass Spectrometry-Based Blood Metabolomics Profiling for Non-Cancer Diseases: A Comprehensive Review. Metabolites 2024; 14:54. [PMID: 38248857 PMCID: PMC10820779 DOI: 10.3390/metabo14010054] [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: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Blood metabolomics profiling using mass spectrometry has emerged as a powerful approach for investigating non-cancer diseases and understanding their underlying metabolic alterations. Blood, as a readily accessible physiological fluid, contains a diverse repertoire of metabolites derived from various physiological systems. Mass spectrometry offers a universal and precise analytical platform for the comprehensive analysis of blood metabolites, encompassing proteins, lipids, peptides, glycans, and immunoglobulins. In this comprehensive review, we present an overview of the research landscape in mass spectrometry-based blood metabolomics profiling. While the field of metabolomics research is primarily focused on cancer, this review specifically highlights studies related to non-cancer diseases, aiming to bring attention to valuable research that often remains overshadowed. Employing natural language processing methods, we processed 507 articles to provide insights into the application of metabolomic studies for specific diseases and physiological systems. The review encompasses a wide range of non-cancer diseases, with emphasis on cardiovascular disease, reproductive disease, diabetes, inflammation, and immunodeficiency states. By analyzing blood samples, researchers gain valuable insights into the metabolic perturbations associated with these diseases, potentially leading to the identification of novel biomarkers and the development of personalized therapeutic approaches. Furthermore, we provide a comprehensive overview of various mass spectrometry approaches utilized in blood metabolomics research, including GC-MS, LC-MS, and others discussing their advantages and limitations. To enhance the scope, we propose including recent review articles supporting the applicability of GC×GC-MS for metabolomics-based studies. This addition will contribute to a more exhaustive understanding of the available analytical techniques. The Integration of mass spectrometry-based blood profiling into clinical practice holds promise for improving disease diagnosis, treatment monitoring, and patient outcomes. By unraveling the complex metabolic alterations associated with non-cancer diseases, researchers and healthcare professionals can pave the way for precision medicine and personalized therapeutic interventions. Continuous advancements in mass spectrometry technology and data analysis methods will further enhance the potential of blood metabolomics profiling in non-cancer diseases, facilitating its translation from the laboratory to routine clinical application.
Collapse
Affiliation(s)
- Ekaterina Demicheva
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Vladislav Dordiuk
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
| | - Fernando Polanco Espino
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
| | - Konstantin Ushenin
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Autonomous Non-Profit Organization Artificial Intelligence Research Institute (AIRI), Moscow 105064, Russia
| | - Saied Aboushanab
- Institute of Chemical Engineering, Ural Federal University, Ekaterinburg 620002, Russia; (S.A.); (V.S.); (E.K.)
| | - Vadim Shevyrin
- Institute of Chemical Engineering, Ural Federal University, Ekaterinburg 620002, Russia; (S.A.); (V.S.); (E.K.)
| | - Aleksey Buhler
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
| | - Elena Mukhlynina
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Olga Solovyova
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Irina Danilova
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Elena Kovaleva
- Institute of Chemical Engineering, Ural Federal University, Ekaterinburg 620002, Russia; (S.A.); (V.S.); (E.K.)
| |
Collapse
|
18
|
Leach ST. Role of Probiotics and Prebiotics in Gut Symbiosis. Nutrients 2024; 16:238. [PMID: 38257131 PMCID: PMC10819279 DOI: 10.3390/nu16020238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 01/24/2024] Open
Abstract
The objective of this Special Issue entitled 'Role of Probiotics and Prebiotics in Gut Symbiosis' is to publish reviews, clinical trials and experimental studies that focus on probiotics and prebiotics that have a role in influencing disease and promoting gastrointestinal and overall health [...].
Collapse
Affiliation(s)
- Steven T Leach
- Department of Paediatrics, School of Clinical Medicine, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|