1
|
Zhang F, Chen Z, Shi J, Han C, Zhan Q, Ren Z, Yang X. - Invited Review - Challenges and constraints to the sustainability of poultry farming in China. Anim Biosci 2025; 38:789-801. [PMID: 39999795 PMCID: PMC11969232 DOI: 10.5713/ab.24.0794] [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: 11/14/2024] [Revised: 12/05/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025] Open
Abstract
China's poultry industry is characterized by large-scale production and rich breeds, presenting both opportunities and challenges. In 2023, the industry produced 10.79 billion broilers, 28.38 million tons of eggs, and 4.88 billion waterfowl. The foundation of a thriving poultry industry lies in the continuous improvement of breeds. For instance, new lines of Lueyang black-boned chickens have been developed using genomic selection breeding, with a focus on improving production performance and unlocking their high-quality genetic potential. Precision nutrition programs enhance the expression of poultry's genetic potential and improve feed utilization efficiency. The five-dimensional feed evaluation system and the comprehensive National Feed Database provide formulators with accurate nutritional parameters of feed. Additionally, the concept of "nutrition power" and the "five-ring gold standard" enable researchers to analyze poultry's digestive physiology more effectively. Feeding management plays a crucial role in optimizing genetic potential and the effectiveness of precision nutrition. To further boost production efficiency, smart farming systems have been implemented, incorporating intelligent management of environmental factors, animal parameters, and poultry health tracking. Meanwhile, in order to improve material utilization efficiency across the entire poultry production chain and support the sustainable development of the poultry industry, it is essential to optimize and promote the application of the Poultry-Crop interaction systems. In summary, strengthening fundamental research in poultry, optimizing smart poultry farm platforms, and implementing Poultry-Crop Interacting systems will drive the sustainable development of China's poultry industry.
Collapse
Affiliation(s)
- Fei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| | - Zhuting Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| | - Jiayi Shi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| | - Chenglong Han
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| | - Qinyi Zhan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| | - Zhouzheng Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi,
China
| |
Collapse
|
2
|
Ding Z, Guo T, Tang Q, Hong Y, Lv Z, Lu L, Zhuang W. DEmiRNA-mRNA regulatory network reveals miR-122-5p as a regulatory factor of arginine metabolism in necrotizing enterocolitis. Front Genet 2025; 15:1480431. [PMID: 39911307 PMCID: PMC11794208 DOI: 10.3389/fgene.2024.1480431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/19/2024] [Indexed: 02/07/2025] Open
Abstract
Objective Necrotizing enterocolitis (NEC) is a gastrointestinal emergency with relatively high morbidity and mortality in neonates. The role of microRNAs (miRNAs) in NEC is not yet entirely clear. This study aimed to explore the mechanism of miR-122-5p in NEC. Methods Differentially expressed (DE) miRNAs were sequenced in control and NEC mice. The DEmiRNA-mRNA regulatory network was constructed and the bioinformatics analysis was performed to identify the target mRNAs and potential roles of the DEmiRNAs. The miR-122-5p activation was explored in vitro in the human intestinal epithelial cell (FHs74Int) and rat intestinal epithelial cell (IEC-6). In vivo, mice were transinfected with miR-122-5p inhibitor before the NEC occurred. Mass spectrometry was used to qualify the concentrations of amino acids, and the viability of intestinal stem cell (ISC) was accessed to verify the biological function. Results Preliminarily, 15 miRNAs were found to be differentially expressed between NEC group and control group. Subsequent bioinformatics analysis revealed that miR-122-5p significantly contributes to the arginine metabolism in NEC through the DEmiRNA-mRNA regulatory network, with PRODH2 and ALDH18A1 being identified as its target genes. In vitro, miR-122-5p mimic inhibited the expression of PRODH2 and ALDH18A1 in the FHs74Int cells and IEC-6 cells. In vivo, inhibition of miR-122-5p led to increased expression of PRODH2 and ALDH18A1, along with elevated arginine levels. Following transfection with a miR-122-5p inhibiting adenovirus, the survival rate of NEC mice improved, and intestinal injury was alleviated. Conclusion MiR-122-5p inhibition could impact arginine metabolism by targeting PRODH2 and ALDH18A1, thereby mitigating intestinal injury in NEC.
Collapse
Affiliation(s)
- Zhili Ding
- Department of General Surgery, Affiliated Changzhou Children’s Hospital of Nantong University, Changzhou, Jiangsu, China
| | - Ting Guo
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Tang
- Department of General Surgery, Affiliated Changzhou Children’s Hospital of Nantong University, Changzhou, Jiangsu, China
| | - Yaqiang Hong
- Department of General Surgery, Affiliated Changzhou Children’s Hospital of Nantong University, Changzhou, Jiangsu, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Lu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjun Zhuang
- Department of General Surgery, Affiliated Changzhou Children’s Hospital of Nantong University, Changzhou, Jiangsu, China
| |
Collapse
|
3
|
Xie Y, Yang M, Huang J, Jiang Z. Identification and Characterization of Genes Associated with Intestinal Ischemia-Reperfusion Injury and Oxidative Stress: A Bioinformatics and Experimental Approach Integrating High-Throughput Sequencing, Machine Learning, and Validation. J Inflamm Res 2025; 18:701-722. [PMID: 39835298 PMCID: PMC11745141 DOI: 10.2147/jir.s500360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Purpose Intestinal ischemia-reperfusion injury (IIRI) occurs as a result of temporary blood flow interruption, leading to tissue damage upon reperfusion. Oxidative stress plays a critical role in this process, instigating inflammation and cell death. Identifying and characterizing genes associated with the oxidative stress response can offer valuable insights into potential therapeutic targets for managing IIRI. Patients and Methods The IIRI dataset was sourced from the NCBI Gene Expression Omnibus Database (GEO), while oxidative stress genes were obtained from the Genecards database. Following the acquisition of differentially expressed genes in IIRI, they were cross-linked with oxidative stress genes to yield IIRI oxidative stress related genes (IOSRGs). The least absolute shrinkage and selection operator, as well as the support vector machine with random forest algorithm, were utilized for machine learning. Subsequently, the PPI network was established, and the Degree and MNC algorithms of the Cytohuba plugin were integrated with the genes obtained through the machine learning algorithms to identify hub IOSRGs (HIOSRGs). A mouse IIRI model and ROC curve were employed to verify the accuracy of HIOSRGs. Finally, siRNA was utilized to suppress the expression of HDAC3 in Caco2 cells, and the changes in oxidative stress levels before and after hypoxia-reoxygenation in Caco2 cells were observed. Results A total of 277 OSRGs and 4 HIOSRGs were obtained. Concurrently, in vivo experimental results of IIRI in C57BL/6 mice, and the establishment of ROC curves, reflected the accuracy and specificity of HIOSRGs. The knockdown of HDAC3 in Caco2 cells resulted in increased oxidative stress levels before and after hypoxia-reoxygenation, underscoring the significant role of HDAC3 in IIRI. Conclusion This study elucidates the interplay between oxidative stress genes and IIRI, offering novel insights into the potential pathogenesis of IIRI and medical interventions for IIRI.
Collapse
Affiliation(s)
- Yongguo Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530007, People’s Republic of China
| | - Mingpu Yang
- General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530007, People’s Republic of China
| | - Juanjuan Huang
- Department of Microbiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Zongbin Jiang
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530007, People’s Republic of China
| |
Collapse
|
4
|
Zhang T, Yan M, Chang M, Hou X, Wang F, Song W, Wang Y, Feng K, Yuan Y, Yue T. Integrated transcriptomics and metabolomics reveal the mechanism of intestinal damage upon acute patulin exposure in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116270. [PMID: 38574645 DOI: 10.1016/j.ecoenv.2024.116270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Mycotoxin contamination has become a major food safety issue and greatly threatens human and animal health. Patulin (PAT), a common mycotoxin in the environment, is exposed through the food chain and damages the gastrointestinal tract. However, its mechanism of enterotoxicity at the genetic and metabolic levels remains to be elucidated. Herein, the intestinal histopathological and biochemical indices, transcriptome, and metabolome of C57BL/6 J mice exposed to different doses of PAT were successively assessed, as well as the toxicokinetics of PAT in vivo. The results showed that acute PAT exposure induced damaged villi and crypts, reduced mucus secretion, decreased SOD and GSH-Px activities, and enhanced MPO activity in the small intestine and mild damage in the colon. At the transcriptional level, the genes affected by PAT were dose-dependently altered in the small intestine and fluctuated in the colon. PAT primarily affected inflammation-related signaling pathways and oxidative phosphorylation in the small intestine and immune responses in the colon. At the metabolic level, amino acids decreased, and extensive lipids accumulated in the small intestine and colon. Seven metabolic pathways were jointly affected by PAT in two intestinal sites. Moreover, changes in PAT products and GST activity were detected in the small intestinal tissue but not in the colonic tissue, explaining the different damage degrees of the two sites. Finally, the integrated results collectively explained the toxicological mechanism of PAT, which damaged the small intestine directly and the colon indirectly. These results paint a clear panorama of intestinal changes after PAT exposure and provide valuable information on the exposure risk and toxic mechanism of PAT.
Collapse
Affiliation(s)
- Ting Zhang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Min Yan
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Min Chang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Xiaohui Hou
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Furong Wang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Wei Song
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Yuan Wang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Kewei Feng
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
5
|
Hu Z, Hua X, Mo X, Chang Y, Chen X, Xu Z, Tao M, Hu G, Song J. Inhibition of NETosis via PAD4 alleviated inflammation in giant cell myocarditis. iScience 2023; 26:107162. [PMID: 37534129 PMCID: PMC10391931 DOI: 10.1016/j.isci.2023.107162] [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: 09/25/2022] [Revised: 04/11/2023] [Accepted: 06/13/2023] [Indexed: 08/04/2023] Open
Abstract
Giant cell myocarditis (GCM) is a rare, usually rapidly progressive, and potentially fatal disease. Detailed inflammatory responses remain unknown, in particular the formation of multinucleate giant cells. We performed single-cell RNA sequencing analysis on 15,714 Cd45+ cells extracted from the hearts of GCM rats and normal rats. NETosis has been found to contribute to the GCM process. An inhibitor of NETosis, GSK484, alleviated GCM inflammation in vivo. MPO (a marker of neutrophils) and H3cit (a marker of NETosis) were expressed at higher levels in patients with GCM than in patients with DCM and healthy controls. Imaging mass cytometry analysis revealed that immune cell types within multinucleate giant cells included CD4+ T cells, CD8+ T cells, neutrophils, and macrophages but not B cells. We elucidated the role of NETosis in GCM pathogenesis, which may serve as a potential therapeutic target in the clinic.
Collapse
Affiliation(s)
- Zhan Hu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Xiuxue Mo
- School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Zhenyu Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Pathology Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Mengtao Tao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Gang Hu
- School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| |
Collapse
|
6
|
Guo T, Hu S, Xu W, Zhou J, Chen F, Gao T, Qu W, Chen F, Lv Z, Lu L. Elevated expression of histone deacetylase HDAC8 suppresses arginine-proline metabolism in necrotizing enterocolitis. iScience 2023; 26:106882. [PMID: 37260741 PMCID: PMC10227426 DOI: 10.1016/j.isci.2023.106882] [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: 08/25/2022] [Revised: 01/07/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
Epigenetic alterations are especially important in necrotizing enterocolitis (NEC). Here, we reported that histone deacetylase 8 (HDAC8) plays a previously unknown role in modulating arginine metabolism via acetylation of histone 3 lysine 9 (acetyl-H3K9) regulation during the pathogenesis of NEC. We found that HDAC8 was upregulated in humans and mice intestinal samples with NEC, while selective inhibition of HDAC8 expression ameliorated NEC. HDAC8 regulates enzymes involved in the metabolic conversion of proline to arginine (PRODH, PRODH2, OAT, and OTC) and arginine to ornithine (ARG1). The results showed that H3K9ac signal in the PRODH/PRODH2 promoter region was mediated by HDAC8. Additionally, the decreased concentration of butyric acid was strongly correlated with elevated HDAC8 levels and circulating arginine, which may result from an unbalanced Firmicutes/Bacteroidetes ratio. These results reveal previously underappreciated roles of microbial metabolites and HDAC8 to coordinate the arginine metabolism during NEC development.
Collapse
Affiliation(s)
- Ting Guo
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Shaohua Hu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Weijue Xu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Jin Zhou
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Feng Chen
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Tingting Gao
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Wenqian Qu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Faling Chen
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Li Lu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| |
Collapse
|
7
|
Nüse B, Holland T, Rauh M, Gerlach RG, Mattner J. L-arginine metabolism as pivotal interface of mutual host-microbe interactions in the gut. Gut Microbes 2023; 15:2222961. [PMID: 37358082 PMCID: PMC10294761 DOI: 10.1080/19490976.2023.2222961] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
L-arginine (L-arg) is a versatile amino acid and a central intestinal metabolite in mammalian and microbial organisms. Thus, L-arg participates as precursor of multiple metabolic pathways in the regulation of cell division and growth. It also serves as a source of carbon, nitrogen, and energy or as a substrate for protein synthesis. Consequently, L-arg can simultaneously modify mammalian immune functions, intraluminal metabolism, intestinal microbiota, and microbial pathogenesis. While dietary intake, protein turnover or de novo synthesis usually supply L-arg in sufficient amounts, the expression of several key enzymes of L-arg metabolism can change rapidly and dramatically following inflammation, sepsis, or injury. Consequently, the availability of L-arg can be restricted due to increased catabolism, transforming L-arg into an essential amino acid. Here, we review the enzymatic pathways of L-arg metabolism in microbial and mammalian cells and their role in immune function, intraluminal metabolism, colonization resistance, and microbial pathogenesis in the gut.
Collapse
Affiliation(s)
- Björn Nüse
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tim Holland
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roman G. Gerlach
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAUErlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Liu K, Tao J, Yang J, Li Y, Su Y, Mao J. Effectiveness of feeding supplementation in preterm infants: an overview of systematic reviews. BMC Pediatr 2022; 22:20. [PMID: 34983444 PMCID: PMC8725413 DOI: 10.1186/s12887-021-03052-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/03/2021] [Indexed: 11/30/2022] Open
Abstract
Background Preterm infants have higher nutrition needs than term infants. The effectiveness of various feeding supplementation was assessed by the improvement of health outcomes in single specific systematic reviews (SRs). The aim of this review was to comprehensively describe the effectiveness of feeding supplementation in promoting health outcomes of preterm infants. Methods A literature search was conducted in the PUBMED, EMBASE, Science Direct, Cochrane library, Web of Science, and Wiley online library. SRs selection followed clear inclusion and exclusion criteria. Pairs of reviewers independently applied the criteria to both titles/abstracts and full texts. Screening and data extraction were performed by using the advanced tables. The methodological quality of SRs and the quality of the evidence were carried out according to the Assessing the Methodological Quality of Systematic Reviews (AMSTAR) tool and the Grades of Recommendation, Assessment, Development, and Evaluation guidelines (GRADE) respectively. A qualitative synthesis of evidence is presented. Results Seventeen SRs were included in the review. Fifteen kinds of feeding supplementation were reported in the SRs. In preterm infants, the effectiveness of feeding supplementation in addition to regular breast-feeding was mainly shown in six aspects: physical health, neurodevelopment, biochemical outcomes, other health outcomes, morbidity and all-cause mortality. And the effectiveness of the interventions on health outcomes in preterm infants was found by most systematic reviews. The methodological quality of all the included SRs was high, and most of the evidences was of low or very low quality. Conclusions Our results will allow a better understanding of the feeding supplementation in preterm infants. Although the feeling supplements may improve the health outcomes of in preterm infants, the existing evidence is uncertain. Therefore, the clinical use of these supplements should be considered cautiously and more well-designed RCTs are still needed to further address the unsolved problems of the included SRs.
Collapse
Affiliation(s)
- Keqin Liu
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jiaxin Tao
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jixin Yang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yufeng Li
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanwei Su
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Jing Mao
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| |
Collapse
|
9
|
Wu YZ, Chan KYY, Leung KT, Lam HS, Tam YH, Lee KH, Li K, Ng PC. The miR-223/nuclear factor I-A axis regulates inflammation and cellular functions in intestinal tissues with necrotizing enterocolitis. FEBS Open Bio 2021; 11:1907-1920. [PMID: 33932136 PMCID: PMC8255851 DOI: 10.1002/2211-5463.13164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
We previously demonstrated that microRNA(miR)‐223 is overexpressed in intestinal tissue of infants with necrotizing enterocolitis (NEC). The objective of the current study was to identify the target gene of miR‐223 and to investigate the role of the miR‐223/nuclear factor I‐A (NFIA) axis in cellular functions that underpin the pathophysiology of NEC. The target gene of miR‐223 was identified by in silico target prediction bioinformatics, luciferase assay, and western blotting. We investigated downstream signals of miR‐223 and cellular functions by overexpressing the miRNA in Caco‐2 and FHs74 cells stimulated with lipopolysaccharide or lipoteichoic acid (LTA). NFIA was identified as a target gene of miR‐223. Overexpression of miR‐223 significantly induced MYOM1 and inhibited NFIA and RGN in Caco‐2 cells, while costimulation with LTA decreased expression of GNA11, MYLK, and PRKCZ. Expression levels of GNA11, MYLK, IL‐6, and IL‐8 were increased, and levels of NFIA and RGN were decreased in FHs74 cells. These potential downstream genes were significantly correlated with levels of miR‐223 or NFIA in primary NEC tissues. Overexpression of miR‐223 significantly increased apoptosis of Caco‐2 and FHs74 cells, while proliferation of FHs74 was inhibited. These results suggest that upon binding with NFIA, miR‐223 regulates functional effectors in pathways of apoptosis, cell proliferation, G protein signaling, inflammation, and smooth muscle contraction. The miR‐223/NFIA axis may play an important role in the pathophysiology of NEC by enhancing inflammation and tissue damage.
Collapse
Affiliation(s)
- Yu Zheng Wu
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kathy Yuen Yee Chan
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kam Tong Leung
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Hugh Simon Lam
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Yuk Him Tam
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kim Hung Lee
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Karen Li
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Pak Cheung Ng
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
10
|
He-Yang J, Zhang W, Liu J, Xue P, Zhou X. Human breast milk oligosaccharides attenuate necrotizing enterocolitis in rats by suppressing mast cell accumulation, DPPI activity and TLR4 expression in ileum tissue, and regulating mitochondrial damage of Caco-2 cells. Int Immunopharmacol 2020; 88:106881. [PMID: 32795899 DOI: 10.1016/j.intimp.2020.106881] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/09/2022]
Abstract
Necrotizing enterocolitis (NEC), a devastating infant disease characterized by severe intestinal necrosis, its pathogenesis is poorly understood, but appears to be multifactorial and highly associated with immaturity of gastrointestinal tract and immature innate-immune system. Breast-milk is effective strategy to protect infants against NEC. This study is using a NEC rat model to investigate the pathological mechanism of NEC involved intestinal-damages, and the therapeutic mechanism of sialylated human milk oligosaccharides (SHMOs) on NEC rats; also using cell model to investigate the effects of SHMOs on colon-epithelial cells (Caco-2) in-vitro. Extraction and characterization of SHMOs from breast milk, establishment of a NEC rat model, histopathological analysis and mast cell accounting of the terminal ileum were taken; The levels of DPPI, TLR4, IL-6, TNF-α, MMP-2/9 and glutathione were measured using various methods. Caco-2 cells were pre-treated with SHMOs and cultured with LPS, histamine, chymase or DPPI, cell viabilities and mitochondrial membrane potential were examined; flow cytometry was used to detect cell cycle. The accumulation of mast cells was found in the ileum of NEC rats, but prohibited by SHMOs treatment; the increased levels of TLR4, DPPI, IL-6, TNF-α, MMP-2/9 in NEC ileum were suppressed by SHMOs in-vivo. SHMOs prevented Caco-2 cells from LPS, histamine, chymase induced damages by surviving cell viability, regulating G0/G1 and S phase in cell cycles, and increasing mitochondrial membrane potential. These findings provide a new insight into the pharmacological mechanism of SHMOs treatment for NEC and suggest that SHMOs needs well attention for therapeutic aims.
Collapse
Affiliation(s)
- Jingqiu He-Yang
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China
| | - Wenting Zhang
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China; Department of Pharmacy, Affiliated Changzhou Children's Hospital of Nantong University, Yanling Road 468, Changzhou 213003, China
| | - Jie Liu
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China
| | - Peng Xue
- Department of Pharmacy, Affiliated Changzhou Children's Hospital of Nantong University, Yanling Road 468, Changzhou 213003, China
| | - Xiaoying Zhou
- The School of Pharmacy, Changzhou University, Jiangsu 213164, China; The School of Medicine, the University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
11
|
Franceschelli S, Gatta DMP, Pesce M, Ferrone A, Quiles JL, Genovese S, Epifano F, Fiorito S, Taddeo VA, Patruno A, Grilli A, Felaco M, Speranza L. Modulation of CAT-2B-Mediated l-Arginine Uptake and Nitric Oxide Biosynthesis in HCT116 Cell Line Through Biological Activity of 4'-Geranyloxyferulic Acid Extract from Quinoa Seeds. Int J Mol Sci 2019; 20:ijms20133262. [PMID: 31269760 PMCID: PMC6650945 DOI: 10.3390/ijms20133262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/27/2019] [Accepted: 06/30/2019] [Indexed: 12/14/2022] Open
Abstract
Chenopodium quinoa Wild is a “pseudocereal” grain which attracts a lot of attention in the scientific community as it has a positive effect on health. Here, we investigate the presence of biologically active O-prenylated phenylpropanoids in the ethanol extract of commercially available quinoa seeds. We claim that 4′-Geranyloxyferulic acid (GOFA) was the only phytochemical product found that belongs to quinoa’s group secondary metabolites. We studied the changes in the oxidative and inflammatory status of the cellular environment in HCT 116 cell line processed with quinoa extract and its component GOFA; the implementation was done through the analysis of the antioxidant enzymes (SOD and CAT), the pro-inflammatory components (iNOS, IL-6 and TNF-α), and the products of intermediary metabolism (ONOO−, O2−). Moreover, the l-arginine uptake was proposed as a target of the tested compounds. We demonstrated that the GOFA, through a decrease of the CAT-2B expression, leads to a reduction of the l-arginine uptake, downregulating the harmful iNOS and restoring the altered redox state. These results propose a new molecular target involved in the reduction of the critical inflammatory process responsible for the cancer progression.
Collapse
Affiliation(s)
- Sara Franceschelli
- Department of Psychological, Health and Territorial Sciences, University G. D'Annunzio, 66100 Chieti, Italy.
| | | | - Mirko Pesce
- Department of Psychological, Health and Territorial Sciences, University G. D'Annunzio, 66100 Chieti, Italy
| | - Alessio Ferrone
- Department of Medicine and Science of Aging, University G. D'Annunzio, 66100 Chieti, Italy
| | - José Luis Quiles
- Department of Physiology, Institute of Nutrition and Food Technology José Mataix, Biomedical Research Centre, University of Granada, 18071 Granada, Spain
| | - Salvatore Genovese
- Department of Pharmacy, University Gabriele D'Annunzio of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo (CH), Italy
| | - Francesco Epifano
- Department of Pharmacy, University Gabriele D'Annunzio of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo (CH), Italy
| | - Serena Fiorito
- Department of Pharmacy, University Gabriele D'Annunzio of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo (CH), Italy
| | - Vito Alessandro Taddeo
- Department of Pharmacy, University Gabriele D'Annunzio of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo (CH), Italy
| | - Antonia Patruno
- Department of Medicine and Science of Aging, University G. D'Annunzio, 66100 Chieti, Italy
| | - Alfredo Grilli
- Department of Psychological, Health and Territorial Sciences, University G. D'Annunzio, 66100 Chieti, Italy
| | - Mario Felaco
- Department of Medicine and Science of Aging, University G. D'Annunzio, 66100 Chieti, Italy
| | - Lorenza Speranza
- Department of Medicine and Science of Aging, University G. D'Annunzio, 66100 Chieti, Italy.
| |
Collapse
|
12
|
Thomaidou A, Chatziioannou AC, Deda O, Benaki D, Gika H, Mikros E, Agakidis C, Raikos N, Theodoridis G, Sarafidis K. A pilot case-control study of urine metabolomics in preterm neonates with necrotizing enterocolitis. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1117:10-21. [PMID: 30991202 DOI: 10.1016/j.jchromb.2019.04.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/01/2019] [Accepted: 04/07/2019] [Indexed: 12/27/2022]
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of gastrointestinal morbidity and mortality in preterm neonates. The aim of this pilot study was to explore using metabolomics alternations in the urine metabolites related to NEC that could possibly serve as diagnostic biomarkers of the disease. Urine samples were prospectively collected at the day of initial evaluation for NEC from 15 diseased preterm neonates (five Bell's stage I and ten stage II/III) and an equal number of matched controls. Urine metabolic profiles were assessed using non-targeted nuclear magnetic resonance spectroscopy and targeted liquid chromatography-tandem mass spectrometry monitoring 108 metabolites. Multivariate statistical models with data from either analytical approach showed clear separation between the metabolic profiles of neonates with NEC and controls. Twenty-five discriminant metabolites were identified belonging to amino and organic acids, sugars and vitamins. A number of metabolite combinations were found to have an excellent diagnostic performance in detecting neonates developing NEC. Our results show that the metabolic profile of neonates with NEC differs significantly from that of controls, making possible their separation using urine metabolomic analysis. Nevertheless, whether the small set of significant metabolites detected in this investigation could be used as early diagnostic biomarkers of NEC should be validated in larger studies.
Collapse
Affiliation(s)
- Agathi Thomaidou
- 1(st) Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, Greece
| | | | - Olga Deda
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece; Biomic_AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, B1.4, Thessaloniki, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001, Greece
| | - Dimitra Benaki
- School of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Helen Gika
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece; Biomic_AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, B1.4, Thessaloniki, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001, Greece
| | - Emmanouel Mikros
- School of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Charalampos Agakidis
- 1(st) Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, Greece
| | - Nikolaos Raikos
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece; Biomic_AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, B1.4, Thessaloniki, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001, Greece
| | - Georgios Theodoridis
- School of Chemistry, Aristotle University of Thessaloniki, Greece; Biomic_AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, B1.4, Thessaloniki, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001, Greece
| | - Kosmas Sarafidis
- 1(st) Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, Greece; Biomic_AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, B1.4, Thessaloniki, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001, Greece.
| |
Collapse
|
13
|
Bazacliu C, Neu J. Pathophysiology of Necrotizing Enterocolitis: An Update. Curr Pediatr Rev 2019; 15:68-87. [PMID: 30387398 DOI: 10.2174/1573396314666181102123030] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/12/2022]
Abstract
NEC is a devastating disease that, once present, is very difficult to treat. In the absence of an etiologic treatment, preventive measures are required. Advances in decoding the pathophysiology of NEC are being made but a more comprehensive understanding is needed for the targeting of preventative strategies. A better definition of the disease as well as diagnostic criteria are needed to be able to specifically label a disease as NEC. Multiple environmental factors combined with host susceptibility appear to contribute to enhanced risks for developing this disease. Several different proximal pathways are involved, all leading to a common undesired outcome: Intestinal necrosis. The most common form of this disease appears to involve inflammatory pathways that are closely meshed with the intestinal microbiota, where a dysbiosis may result in dysregulated inflammation. The organisms present in the intestinal tract prior to the onset of NEC along with their diversity and functional capabilities are just beginning to be understood. Fulfillment of postulates that support causality for particular microorganisms is needed if bacteriotherapies are to be intelligently applied for the prevention of NEC. Identification of molecular effector pathways that propagate inflammation, understanding of, even incipient role of genetic predisposition and of miRNAs may help solve the puzzle of this disease and may bring the researchers closer to finding a treatment. Despite recent progress, multiple limitations of the current animal models, difficulties related to studies in humans, along with the lack of a "clear" definition will continue to make it a very challenging disease to decipher.
Collapse
Affiliation(s)
- Catalina Bazacliu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| |
Collapse
|
14
|
Plasma citrulline is a sensitive safety biomarker for small intestinal injury in rats. Toxicol Lett 2018; 295:416-423. [DOI: 10.1016/j.toxlet.2018.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/22/2018] [Accepted: 07/06/2018] [Indexed: 11/21/2022]
|
15
|
Robinson JL, Smith VA, Stoll B, Agarwal U, Premkumar MH, Lau P, Cruz SM, Manjarin R, Olutoye O, Burrin DG, Marini JC. Prematurity reduces citrulline-arginine-nitric oxide production and precedes the onset of necrotizing enterocolitis in piglets. Am J Physiol Gastrointest Liver Physiol 2018; 315:G638-G649. [PMID: 30048597 PMCID: PMC6415715 DOI: 10.1152/ajpgi.00198.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis (NEC) is associated with low plasma arginine and vascular dysfunction. It is not clear whether low intestinal citrulline production, the precursor for arginine synthesis, occurs before and thus predisposes to NEC or if it results from tissue damage. This study was designed to test the hypothesis that whole body rates of citrulline, arginine, and nitric oxide synthesis are low in premature pigs and that they precede NEC. Piglets delivered by cesarean section at 103 days [preterm (PT)], 110 days [near-term (NT)], or 114 days [full-term (FT)] of gestation were given total parenteral nutrition and after 2 days orogastrically fed infant formula for 42 h to induce NEC. Citrulline and arginine fluxes were determined before and during the feeding protocol. Gross macroscopic and histological NEC scores and plasma fatty acid binding protein (iFABP) concentration were determined as indicators of NEC. Intestinal gene expression for enzymes of the arginine pathway were quantitated. A lower ( P < 0.05) survival rate was observed for PT (8/27) than for NT (9/9) and FT pigs (11/11). PT pigs had higher macroscopic gross ( P < 0.05) and histological NEC ( P < 0.05) scores and iFABP concentration ( P < 0.05) than pigs of more advanced gestational age. PT pigs had lower citrulline production and arginine fluxes ( P < 0.05) throughout and a reduced gene expression in genes of the citrulline-arginine pathway. In summary, intestinal enzyme expression and whole body citrulline and arginine fluxes were reduced in PT pigs compared with animals of more advance gestational age and preceded the development of NEC. NEW & NOTEWORTHY Arginine supplementation prevents necrotizing enterocolitis (NEC), the most common gastrointestinal emergency of prematurity. Citrulline (precursor for arginine) production is reduced during NEC, and this is believed to be a consequence of intestinal damage. In a swine model of NEC, we show that intestinal gene expression of the enzymes for citrulline production and whole body citrulline and arginine fluxes are reduced and precede the onset of NEC in premature pigs. Reduced citrulline production during prematurity may be a predisposition to NEC.
Collapse
Affiliation(s)
- Jason L. Robinson
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Victoria A. Smith
- 2Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Barbara Stoll
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Umang Agarwal
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Muralidhar H. Premkumar
- 3Division of Neonatology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Patricio Lau
- 4Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Stephanie M. Cruz
- 4Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Rodrigo Manjarin
- 2Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Oluyinka Olutoye
- 4Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Douglas G. Burrin
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Juan C. Marini
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas,5Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|