1
|
He X, Wei W, Liu J, Liang Z, Wu Y, Liu J, Pi J, Zhang H. Whole-transcriptome analysis reveals the effect of retinoic acid on small intestinal mucosal injury in cage-stressed young laying ducks. Poult Sci 2024; 103:104376. [PMID: 39423790 PMCID: PMC11532482 DOI: 10.1016/j.psj.2024.104376] [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: 07/15/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024] Open
Abstract
Retinoic acid (RA) is an active derivative of vitamin A and is involved in a variety of physiological processes, including cell growth, antioxidant, and inflammation. However, the role of RA in intestinal oxidative stress injury in caged-stressed laying ducks is unknown. In this study, we analyzed the effect and underlying mechanism of RA supplementation on intestinal damage in cage-stressed young laying ducks. One hundred and sixty laying ducks were divided into 5 treatment groups, including a control group (CR) and 4 treatment groups exposed to different RA concentrations (2,500, 5,000, 7,500 and 10,000 IU/kg, TG1 to TG4). The experimental period comprised a 7-d prefeeding period and a 10-d experimental feeding period, for a total of 17 d. Phenotypic analysis revealed that compared with the control group, RA addition increased the intestinal villus height and the villus-to-crypt ratio; decreased the crypt depth (P < 0.01); decreased the serum diamine oxidase and D-lactate concentrations (P < 0.05); increased the serum antioxidant capacity and intestinal antioxidant gene expression levels (P < 0.05); and increased the expression levels of tight junction-related genes, with the greatest effect observed in TG2 group. Our further whole-transcriptome analysis of duodenum tissues from CR and TG2 ducks revealed 706 differentially expressed mRNAs (DEmRNAs), 357 differentially expressed lncRNAs (DElncRNAs), 14 differentially expressed circRNAs (DEcircRNAs), and 4 differentially expressed miRNAs (DEmiRNAs). These DEGs are involved in calcium signaling, NOD-like receptor signaling, pyruvate metabolism, Jak-STAT signaling, Wnt signaling, riboflavin metabolism, and the adherens junction and tight junction pathways. The results of omics and marker gene expression analysis suggested that RA treatment may play a role in endoplasmic reticulum stress (ERS) and apoptosis. In conclusion, the addition of RA to the diet improved intestinal injury by improving the redox homeostasis of intestinal cells associated with ERS, enhancing the intestinal tight junction structure and alleviating the apoptosis of intestinal epithelial cells; moreover, 5,000 IU/kg RA was determined to be the most appropriate concentration for supplementation.
Collapse
Affiliation(s)
- Xiaolong He
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Wenzhuo Wei
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Jia Liu
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Zhenhua Liang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Yan Wu
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Jinsong Pi
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Hao Zhang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.
| |
Collapse
|
2
|
Zhang Y, Luo Y, Shi J, Xie Y, Shao H, Li Y. All-trans retinoic acid alleviates collagen-induced arthritis and promotes intestinal homeostasis. Sci Rep 2024; 14:1811. [PMID: 38245637 PMCID: PMC10799902 DOI: 10.1038/s41598-024-52322-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Abstract
All-trans retinoic acid (ATRA) has emerged as a promising adjunctive treatment for rheumatoid arthritis. However, the mechanism by which ATRA mitigates arthritis remains unclear. In this study, we aimed to explore ATRA alleviation of arthritis and the role of ATRA in regulating intestinal homeostasis. Thus, we established a collagen-induced arthritis (CIA) model in Wistar rats. After 6 weeks of ATRA treatment, the arthritis index of CIA rats decreased, synovial inflammation was alleviated, and the disruption of Th17/Treg differentiation in peripheral blood was reversed. Additionally, the Th17/Treg ratio in the mesenteric lymph nodes decreased and the expression of Foxp3 mRNA increased and that of IL-17 mRNA decreased in the colon and ileum. Microscopically, we observed reduced intestinal inflammation. Transmission electron microscopy revealed that ATRA could repair tight junctions, which was accompanied by an increase in the expression of Claudin-1, Occludin and ZO-1. Moreover, ATRA regulated the composition of the gut microbiota, as was characterized based on the reduced abundance of Desulfobacterota and the increased abundance of Lactobacillus. In conclusion, ATRA demonstrates the potential to alleviate arthritis in CIA rats, which might be correlated with modulating the gut microbiota and regulating the intestinal immune response. Our findings provide novel insights into ATRA-mediated alleviation of arthritis.
Collapse
Affiliation(s)
- Yiqi Zhang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yating Luo
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiangchun Shi
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yumeng Xie
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huangfang Shao
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
- Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Yang J, Chen X, Liu T, Shi Y. Potential role of bile acids in the pathogenesis of necrotizing enterocolitis. Life Sci 2024; 336:122279. [PMID: 37995935 DOI: 10.1016/j.lfs.2023.122279] [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: 09/01/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most common acute gastrointestinal diseases in preterm infants. Recent studies have found that NEC is not only caused by changes in the intestinal environment but also by the failure of multiple systems and organs, including the liver. The accumulation of bile acids (BAs) in the ileum and the disorder of ileal BA transporters are related to the ileum injury of NEC. Inflammatory factors such as tumor necrosis factor (TNF)-α and interleukin (IL)-18 secreted by NEC also play an important role in regulating intrahepatic BA transporters. As an important link connecting the liver and intestinal circulation, the bile acid metabolic pathway plays an important role in the regulation of intestinal microbiota, cell proliferation, and barrier protection. In this review, we focus on how bile acids explore the dynamic changes of bile acid metabolism in necrotizing enterocolitis and the potential therapeutic value of targeting the bile acid signaling pathways.
Collapse
Affiliation(s)
- Jiahui Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tianjing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
4
|
Zhang L, Tang R, Wu Y, Liang Z, Liu J, Pi J, Zhang H. The Role and Mechanism of Retinol and Its Transformation Product, Retinoic Acid, in Modulating Oxidative Stress-Induced Damage to the Duck Intestinal Epithelial Barrier In Vitro. Animals (Basel) 2023; 13:3098. [PMID: 37835704 PMCID: PMC10572057 DOI: 10.3390/ani13193098] [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: 08/24/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
This study aimed to investigate the effects and mechanisms of retinol and retinoic acid on primary duck intestinal epithelial cells under oxidative stress induced by H2O2. Different ratios of retinol and retinoic acid were used for treatment. The study evaluated the cell morphology, viability, antioxidative capacity, and barrier function of cells. The expression of genes related to oxidative stress and the intestinal barrier was analyzed. The main findings demonstrated that the treated duck intestinal epithelial cells exhibited increased viability, increased antioxidative capacity, and improved intestinal barrier function compared to the control group. High retinoic acid treatment improved viability and gene expression, while high retinol increased antioxidative indicators and promoted intestinal barrier repair. Transcriptome analysis revealed the effects of treatments on cytokine interactions, retinol metabolism, PPAR signaling, and cell adhesion. In conclusion, this study highlights the potential of retinol and retinoic acid in protecting and improving intestinal cell health under oxidative stress, providing valuable insights for future research.
Collapse
Affiliation(s)
- Li Zhang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (R.T.); (Y.W.); (Z.L.); (J.P.)
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Rui Tang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (R.T.); (Y.W.); (Z.L.); (J.P.)
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Yan Wu
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| | - Zhenhua Liang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Jinsong Pi
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| | - Hao Zhang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (R.T.); (Y.W.); (Z.L.); (J.P.)
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| |
Collapse
|
5
|
Kim HW, Lee SY, Hur SJ, Kil DY, Kim JH. Effects of functional nutrients on chicken intestinal epithelial cells induced with oxidative stress. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2023; 65:1040-1052. [PMID: 37969347 PMCID: PMC10640939 DOI: 10.5187/jast.2023.e22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 11/17/2023]
Abstract
The objective of this study was to investigate the protective effects of functional nutrients including various functional amino acids, vitamins, and minerals on chicken intestinal epithelial cells (cIECs) treated with oxidative stress. The cIECs were isolated from specific pathogen free eggs. Cells were exposed to 0 mM supplement (control), 20 mM threonine (Thr), 0.4 mM tryptophan (Trp), 1 mM glycine (Gly), 10 μM vitamin C (VC), 40 μM vitamin E (VE), 5 μM vitamin A (VA), 34 μM chromium (Cr), 0.42 μM selenium (Se), and 50 μM zinc (Zn) for 24 h with 6 replicates for each treatment. After 24 h, cells were further incubated with fresh culture medium (positive control, PC) or 1 mM H2O2 with different supplements (negative control, NC and each treatment). Oxidative stress was measured by cell proliferation, whereas tight junction barrier function was analyzed by fluorescein isothiocyanate (FITC)-dextran permeability and transepithelial electrical resistance (TEER). Results indicated that cell viability and TEER values were less (p < 0.05) in NC treatments with oxidative stress than in PC treatments. In addition, FITC-dextran values were greater (p < 0.05) in NC treatments with oxidative stress than in PC treatments. The supplementations of Thr, Trp, Gly, VC, and VE in cells treated with H2O2 showed greater (p < 0.05) cell viability than the supplementation of VA, Cr, Se, and Zn. The supplementations of Trp, Gly, VC, and Se in cells treated with H2O2 showed the least (p < 0.05) cellular permeability. In addition, the supplementation of Thr, VE, VA, Cr, and Zn in cells treated with H2O2 decreased (p < 0.05) cellular permeability. At 48 h, the supplementations of Thr, Trp, and Gly in cells treated with H2O2 showed the greatest (p < 0.05) TEER values among all treatments, and the supplementations of VC and VE in cells treated with H2O2 showed greater (p < 0.05) TEER values than the supplementations of VA, Cr, Se, and Zn in cells treated with H2O2. In conclusion, Thr, Trp, Gly, and VC supplements were effective in improving cell viability and intestinal barrier function of cIECs exposed to oxidative stress.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Seung Yun Lee
- Department of Animal Science (BK21 Four),
Institute of Agriculture Life Science, Gyeongsang National
University, Jinju 52725, Korea
| | - Sun Jin Hur
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Dong Yong Kil
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Jong Hyuk Kim
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| |
Collapse
|
6
|
Luo P, Zheng L, Zou J, Chen T, Zou J, Li W, Chen Q, Qian B. Insights into vitamin A in bladder cancer, lack of attention to gut microbiota? Front Immunol 2023; 14:1252616. [PMID: 37711628 PMCID: PMC10497765 DOI: 10.3389/fimmu.2023.1252616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Vitamin A has long been associated with bladder cancer, and many exogenous vitamin A supplements, vitamin A derivatives, and synthetic drugs have been investigated over the years. However, the effectiveness of these strategies in clinical practice has not met expectations, and they have not been widely adopted. Recent medical research on intestinal flora has revealed that bladder cancer patients exhibit reduced serum vitamin A levels and an imbalance of gut microbiota. In light of the close relationship between gut microbiota and vitamin A, one can speculate that a complex regulatory mechanism exists between the two in the development and occurrence of bladder cancer. As such, further exploration of their interaction in bladder cancer may help guide the use of vitamin A for preventive purposes. During the course of this review, attention is paid to the influence of intestinal microbiota on the vitamin A metabolism and the RA signaling pathway, as well as the mutual promotion relationships between them in the prevention of bladder cancer, In addition, it emphasizes the importance of intestinal microbiota for bladder cancer prevention and treatment.
Collapse
Affiliation(s)
- Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Jun Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| |
Collapse
|
7
|
Yao S, Zhao Y, Chen H, Sun R, Chen L, Huang J, Yu Z, Chen S. Exploring the Plasticity of Diet on Gut Microbiota and Its Correlation with Gut Health. Nutrients 2023; 15:3460. [PMID: 37571397 PMCID: PMC10420685 DOI: 10.3390/nu15153460] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Dietary habits have been proven to help alter the composition of gut microbiota, and exploring the impact of nutritional patterns on gut microbiota changes can help protect gut health. However, few studies have focused on the dietary impact on the gut microbiota over an experimental timeframe. In this study, 16S rRNA gene sequencing was employed to investigate the gut microbiota of mice under different dietary patterns, including AIN-93G diet (Control), high protein diet (HPD), high fiber diet (HFD), and switch diet (Switch). The alpha diversity of the HPD group significantly decreased, but HFD can restore this decline. During HPD, some genera were significantly upregulated (e.g., Feacalibaculum) and downregulated (e.g., Parabacteroides). However, after receiving HFD, other genera were upregulated (e.g., Akkermansia) and downregulated (e.g., Lactobacillus). In addition, the interaction between pathogenic bacteria was more pronounced during HPD, while the main effect was probiotics during HFD. In conclusion, the plasticity exhibited by the gut microbiota was subject to dietary influences, wherein disparate dietary regimens hold pivotal significance in upholding the well-being of the host. Therefore, our findings provide new ideas and references for the relationship between diets and gut microbiota.
Collapse
Affiliation(s)
- Siqi Yao
- Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha 410008, China;
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Yiming Zhao
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Hao Chen
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410078, China; (H.C.); (J.H.)
| | - Ruizheng Sun
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Liyu Chen
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410078, China; (H.C.); (J.H.)
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Shuijiao Chen
- Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410008, China
| |
Collapse
|
8
|
Sami AS, Frazer LC, Miller CM, Singh DK, Clodfelter LG, Orgel KA, Good M. The role of human milk nutrients in preventing necrotizing enterocolitis. Front Pediatr 2023; 11:1188050. [PMID: 37334221 PMCID: PMC10272619 DOI: 10.3389/fped.2023.1188050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an intestinal disease that primarily impacts preterm infants. The pathophysiology of NEC involves a complex interplay of factors that result in a deleterious immune response, injury to the intestinal mucosa, and in its most severe form, irreversible intestinal necrosis. Treatments for NEC remain limited, but one of the most effective preventative strategies for NEC is the provision of breast milk feeds. In this review, we discuss mechanisms by which bioactive nutrients in breast milk impact neonatal intestinal physiology and the development of NEC. We also review experimental models of NEC that have been used to study the role of breast milk components in disease pathophysiology. These models are necessary to accelerate mechanistic research and improve outcomes for neonates with NEC.
Collapse
Affiliation(s)
- Ahmad S. Sami
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lynda G. Clodfelter
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
9
|
Shu LZ, Ding YD, Xue QM, Cai W, Deng H. Direct and indirect effects of pathogenic bacteria on the integrity of intestinal barrier. Therap Adv Gastroenterol 2023; 16:17562848231176427. [PMID: 37274298 PMCID: PMC10233627 DOI: 10.1177/17562848231176427] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/01/2023] [Indexed: 06/06/2023] Open
Abstract
Bacterial translocation is a pathological process involving migration of pathogenic bacteria across the intestinal barrier to enter the systemic circulation and gain access to distant organs. This phenomenon has been linked to a diverse range of diseases including inflammatory bowel disease, pancreatitis, and cancer. The intestinal barrier is an innate structure that maintains intestinal homeostasis. Pathogenic infections and dysbiosis can disrupt the integrity of the intestinal barrier, increasing its permeability, and thereby facilitating pathogen translocation. As translocation represents an essential step in pathogenesis, a clear understanding of how barrier integrity is disrupted and how this disruption facilitates bacterial translocation could identify new routes to effective prophylaxis and therapy. In this comprehensive review, we provide an in-depth analysis of bacterial translocation and intestinal barrier function. We discuss currently understood mechanisms of bacterial-enterocyte interactions, with a focus on tight junctions and endocytosis. We also discuss the emerging concept of bidirectional communication between the intestinal microbiota and other body systems. The intestinal tract has established 'axes' with various organs. Among our regulatory systems, the nervous, immune, and endocrine systems have been shown to play pivotal roles in barrier regulation. A mechanistic understanding of intestinal barrier regulation is crucial for the development of personalized management strategies for patients with bacterial translocation-related disorders. Advancing our knowledge of barrier regulation will pave the way for future research in this field and novel clinical intervention strategies.
Collapse
Affiliation(s)
- Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Yi-Dan Ding
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Qing-Ming Xue
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Wei Cai
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
- Department of Pathology, the Fourth Affiliated
Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated
Hospital of Nanchang University, No. 133 South Guangchang Road, Nanchang
330003, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang
University, Nanchang, China
| |
Collapse
|
10
|
Guardiola-Márquez CE, Santos-Ramírez MT, Segura-Jiménez ME, Figueroa-Montes ML, Jacobo-Velázquez DA. Fighting Obesity-Related Micronutrient Deficiencies through Biofortification of Agri-Food Crops with Sustainable Fertilization Practices. PLANTS (BASEL, SWITZERLAND) 2022; 11:3477. [PMID: 36559589 PMCID: PMC9784404 DOI: 10.3390/plants11243477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 06/17/2023]
Abstract
Obesity is a critical medical condition worldwide that is increasingly involved with nutritional derangements associated with micronutrient deficiencies, including iron, zinc, calcium, magnesium, selenium, and vitamins A, C, D, and E. Nutritional deficiencies in obesity are mainly caused by poor-quality diets, higher nutrient requirements, alterations in micronutrient metabolism, and invasive obesity treatments. The current conventional agricultural system is designed for intensive food production, focusing on food quantity rather than food quality, consuming excessive agricultural inputs, and producing nutrient-deficient foods, thus generating severe health and environmental problems; agricultural food products may worsen obesity-related malnutrition. Therefore, modern agriculture is adopting new biofortification technologies to combat micronutrient deficiencies and improve agricultural productivity and sustainability. Biofertilization and nanofertilization practices are increasingly used due to their efficiency, safety, and reduced environmental impact. Biofertilizers are preparations of PGP-microorganisms that promote plant growth by influencing plant metabolism and improving the nutrient uptake, and nanofertilizers consist of synthesized nanoparticles with unique physicochemical properties that are capable of increasing plant nutrition and enriching agricultural products. This review presents the current micronutrient deficiencies associated with obesity, the modern unsustainable agri-food system contributing to obesity progression, and the development of bio- and nanofertilizers capable of biofortifying agri-food crops with micronutrients commonly deficient in patients with obesity.
Collapse
Affiliation(s)
| | - María Teresa Santos-Ramírez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
| | - M. Eugenia Segura-Jiménez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
| | - Melina Lizeth Figueroa-Montes
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
| | - Daniel A. Jacobo-Velázquez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
- Tecnologico de Monterrey, The Institute for Obesity Research, Ave. General Ramon Corona 2514, Zapopan 45201, Jalisco, Mexico
| |
Collapse
|
11
|
Gao F, Zhang L, Li H, Xia F, Bai H, Piao X, Sun Z, Cui H, Shi L. Dietary Oregano Essential Oil Supplementation Influences Production Performance and Gut Microbiota in Late-Phase Laying Hens Fed Wheat-Based Diets. Animals (Basel) 2022; 12:ani12213007. [PMID: 36359131 PMCID: PMC9654440 DOI: 10.3390/ani12213007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
This study aimed to investigate the potential effects of OEO on production performance, egg quality, fatty acid composition in yolk, and cecum microbiota of hens in the late phase of production. A total of 350 58-week-old Jing Tint Six laying hens were randomly divided into five groups: (1) fed a basal diet (control); (2) fed a basal diet + 5 mg/kg flavomycin (AGP); (3) fed a basal diet + 100 mg/kg oregano essential oil + 20 mg/kg cinnamaldehyde (EO1); (4) fed a basal diet + 200 mg/kg oregano essential oil + 20 mg/kg cinnamaldehyde (EO2); (5) fed a basal diet + 300 mg/kg oregano essential oil + 20 mg/kg cinnamaldehyde (EO3). Compared to the control group, group EO2 exhibited higher (p < 0.05) egg production during weeks 5−8 and 1−8. EO2 had a lower feed conversion ratio than the control group during weeks 1−8. The content of monounsaturated fatty acid (MUFA) in EO2 was higher (p < 0.05) than that of the control and AGP groups. EO2 increased (p < 0.05) the abundance of Actinobacteriota and decreased the abundance of Desulfovibri in the cecum. The abundances of Anaerofilum, Fournierella, Fusobacterium, and Sutterella were positively correlated with egg production, feed conversion ratio, and average daily feed intake, while the abundances of Bacteroides, Desulfovibrio, Lactobacillus, Methanobrevibacter, and Rikenellaceae_RC9_gut_group were negatively correlated with egg production, feed conversion ratio, and average daily feed intake. Dietary supplementation with 200 mg/kg OEO and 20 mg/kg cinnamaldehyde could improve egg-production performance, decrease feed conversion ratio, and alter the fatty acid and microbial composition of eggs from late-phase laying hens.
Collapse
Affiliation(s)
- Fei Gao
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lianhua Zhang
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
| | - Hui Li
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
| | - Fei Xia
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
| | - Hongtong Bai
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
| | - Xiangshu Piao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhiying Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hongxia Cui
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
| | - Lei Shi
- Key Laboratory of Plant Resources and Beijing Botanical Garden, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- China National Botanical Garden, Beijing 100093, China
- Correspondence:
| |
Collapse
|
12
|
Yang H, Qu Y, Gao Y, Sun S, Wu R, Wu J. Research Progress on the Correlation between the Intestinal Microbiota and Food Allergy. Foods 2022; 11:foods11182913. [PMID: 36141041 PMCID: PMC9498665 DOI: 10.3390/foods11182913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
The increasing incidence of food allergy is becoming a substantial public health concern. Increasing evidence suggests that alterations in the composition of the intestinal microbiota play a part in the development of food allergy. Additionally, the application of probiotics to correct gut microbiota imbalances and regulate food allergy has become a research hotspot. However, the mechanism by which the gut microbiota regulates food allergy and the efficacy of probiotics are still in the preliminary exploration stage, and there are no clear and specific conclusions. The aim of this review is to provide information regarding the immune mechanism underlying food allergy, the correlation between the intestinal microbiota and food allergy, a detailed description of causation, and mechanisms by which the intestinal microbiota regulates food allergy. Subsequently, we highlight how probiotics modulate the gut microbiome–immune axis to alleviate food allergy. This study will contribute to the dovetailing of bacterial therapeutics with immune system in allergic individuals to prevent food allergy and ameliorate food allergy symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Rina Wu
- Correspondence: or ; Tel./Fax: +86-24-88487161
| | | |
Collapse
|
13
|
Tsugeno Y, Sato T, Watanabe M, Higashide M, Furuhashi M, Umetsu A, Suzuki S, Ida Y, Hikage F, Ohguro H. All Trans-Retinoic Acids Facilitate the Remodeling of 2D and 3D Cultured Human Conjunctival Fibroblasts. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9090463. [PMID: 36135009 PMCID: PMC9495389 DOI: 10.3390/bioengineering9090463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022]
Abstract
Vitamin A derivative, all-trans-retinoic acid (ATRA), is known to be a potent regulator of the growth and differentiation of various types of cells. In the present study, the unidentified effects of ATRA on superficial and vertical spreading conjunctival scarring were examined. The study involved the use of two-dimensional (2D) and three-dimensional (3D) cultures of human conjunctival fibroblast (HconF) cells in the presence or absence of TGF-β2. The effects of ATRA (1 μM) on superficial or vertical spreading conjunctival scarring were evaluated by the barrier function by trans-endothelial electrical resistance (TEER) and FITC dextran permeability measurements and real-time metabolic analysis, as well as the physical properties, namely, the size and stiffness, of 3D spheroids, respectively. In addition, the expressions of several related molecules, including extracellular matrix (ECM) molecules, ECM modulators including a tissue inhibitor of metalloproteinases (TIMPs), matrix metalloproteinases (MMPs), and ER stress-related factors, were examined. ATRA significantly induced (1) an increase in TEER values and a decrease in FITC dextran permeability, respectively, in the 2D monolayers, and (2) relatively and substantially increased the size and stiffness, respectively, of the 3D spheroids. These ATRA-induced effects were further enhanced in the TGF-β2-treated cells, whereas the TGF-β2-induced enhancement in glycolytic capacity was canceled by the presence of ATRA. Consistent with these physical and morphological effects, the mRNA expressions of several molecules were significantly but differently induced between 2D and 3D cultures by ATRA, although the presence of TGF-β2 did not substantially affect these gene expression levels. The findings reported in this study indicate that ATRA may exacerbate both superficial and vertical conjunctival fibrosis spreading independently of TGF-β2-induced changes.
Collapse
Affiliation(s)
- Yuri Tsugeno
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Soma Suzuki
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yosuke Ida
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Fumihito Hikage
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
- Correspondence: ; Tel.: +81-116-112-111; Fax: +81-116-136-575
| |
Collapse
|
14
|
Vitamin–Microbiota Crosstalk in Intestinal Inflammation and Carcinogenesis. Nutrients 2022; 14:nu14163383. [PMID: 36014889 PMCID: PMC9414212 DOI: 10.3390/nu14163383] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CAC) are common diseases of the digestive system. Vitamin deficiencies and gut microbiota dysbiosis have a close relationship with the risk, development, and progression of IBD and CAC. There is a strong link between vitamins and the gut microbiome. Vitamins are extremely crucial for maintaining a healthy gut microbiota, promoting growth and development, metabolism, and innate immunity. Gut microbiota can not only influence the transport process of vitamins, but also produce vitamins to compensate for insufficient food intake. Emerging evidence suggests that oral vitamin supplementation can reduce inflammation levels and improve disease prognosis. In addition, improving the diet structure and consuming foods rich in vitamins not only help to improve the vitamin deficiency, but also help to reduce the risk of IBD. Fecal microbiota transplantation (FMT) and the application of vitamin-producing probiotics can better assist in the treatment of intestinal diseases. In this review, we discuss the interaction and therapeutic roles of vitamins and gut microbiota in IBD and CAC. We also summarize the methods of treating IBD and CAC by modulating vitamins. This may highlight strategies to target gut-microbiota-dependent alterations in vitamin metabolism in the context of IBD and CAC therapy.
Collapse
|
15
|
Berken JA, Chang J. Neurologic consequences of neonatal necrotizing enterocolitis. Dev Neurosci 2022; 44:295-308. [PMID: 35697005 DOI: 10.1159/000525378] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 05/24/2022] [Indexed: 11/19/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease of the premature infant with high mortality and morbidity. Children who survive NEC have been shown to demonstrate neurodevelopmental delay, with significantly worse outcomes than from prematurity alone. The pathways leading to NEC-associated neurological impairments remain unclear, limiting the development of preventative and protective strategies. This review aims to summarize the existing clinical and experimental studies related to NEC-associated brain injury. We describe the current epidemiology of NEC, reported long-term neurodevelopmental outcomes among survivors, and proposed pathogenesis of brain injury in NEC. Highlighted are the potential connections between hypoxia-ischemia, nutrition, infection, gut inflammation, and the developing brain in NEC.
Collapse
Affiliation(s)
- Jonathan A Berken
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Jill Chang
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA,
- Division of Neonatal-Perinatal Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA,
| |
Collapse
|
16
|
Wells JM, Gao Y, de Groot N, Vonk MM, Ulfman L, van Neerven RJJ. Babies, Bugs, and Barriers: Dietary Modulation of Intestinal Barrier Function in Early Life. Annu Rev Nutr 2022; 42:165-200. [PMID: 35697048 DOI: 10.1146/annurev-nutr-122221-103916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The intestinal barrier is essential in early life to prevent infection, inflammation, and food allergies. It consists of microbiota, a mucus layer, an epithelial layer, and the immune system. Microbial metabolites, the mucus, antimicrobial peptides, and secretory immunoglobulin A (sIgA) protect the intestinal mucosa against infection. The complex interplay between these functionalities of the intestinal barrier is crucial in early life by supporting homeostasis, development of the intestinal immune system, and long-term gut health. Exclusive breastfeeding is highly recommended during the first 6 months. When breastfeeding is not possible, milk-based infant formulas are the only safe alternative. Breast milk contains many bioactive components that help to establish the intestinal microbiota and influence the development of the intestinal epithelium and the immune system. Importantly, breastfeeding lowers the risk for intestinal and respiratory tract infections. Here we review all aspects of intestinal barrier function and the nutritional components that impact its functionality in early life, such as micronutrients, bioactive milk proteins, milk lipids, and human milk oligosaccharides. These components are present in breast milk and can be added to milk-based infant formulas to support gut health and immunity. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Jerry M Wells
- Host Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Yifan Gao
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands
| | | | | | | | - R J Joost van Neerven
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands.,FrieslandCampina, Amersfoort, The Netherlands;
| |
Collapse
|
17
|
Zhang Y, Wang O, Mi H, Yi J, Cai S. Rhus chinensis Mill. fruits prevent necrotizing enterocolitis in rat pups via regulating the expressions of key proteins involved in multiple signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115103. [PMID: 35157955 DOI: 10.1016/j.jep.2022.115103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on ancient records and previous studies, many parts of Rhus chinensis Mill., including the fruits, have good preventive and therapeutic effects on inflammation, malaria, diarrhea, and gastrointestinal diseases. Rhus plants and Galla chinensis produced from R. chinensis leaves can also prevent or cure intestinal diseases. However, the preventive effect and molecular mechanisms of R. chinensis fruits on necrotizing enterocolitis (NEC) have not been comprehensively studied. AIM OF THE STUDY This article aims to estimate the effect of the 80% ethanol extract of R. chinensis fruits (RM) on alleviating NEC in rat pups and illustrate the potential molecular mechanisms. MATERIALS AND METHODS Rat pups were subjected to formula feeding, intermittent hypoxic, and cold stresses to establish the NEC model. The preventive effects of RM on NEC were evaluated through survival rate; clinical sickness index; macroscopic conditions; histopathology; and expression levels of inflammatory markers (i.e., tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6]), oxidative stress indicators (i.e., total antioxidant status [TAS], total oxidant status [TOS], superoxide dismutase [SOD], glutathione peroxidase [GSH-Px], myeloperoxidase [MPO], malondialdehyde [MDA]), and tight junction proteins (i.e., Zonula Occludens 1 [ZO-1], Occludin). Moreover, the expression levels of several key proteins involved in oxidative stress (i.e., nuclear factor erythroid 2-related factor 2 [Nrf2], NAD(P)H-quinone oxidoreductase-1 [NQO1]), inflammation (i.e., Toll-like receptor 4 [TLR4], phosphorylated-nuclear factor kappa-B [p-NF-κB], inducible nitric oxide synthase [iNOS]), and apoptosis (i.e., cleaved cysteinyl aspartate specific proteinase-3 [cleaved Caspase-3], Bcl-2-associated X [Bax], B-cell lymphoma-2 [Bcl-2]) in intestinal tissues were analyzed to clarify the molecular mechanisms. RESULTS The extract particularly high doses (400 mg RM/kg body weight) could remarkably reduce the mortality and clinical sickness score and improve the macroscopic condition and histopathological injury of the intestine in NEC pups. After RM administration, the levels of TOS, TNF-α, IL-6, MPO, and MDA in the bowel tissue decreased, whereas the levels of TAS, SOD, and GSH-Px were significantly enhanced. The expression levels of ZO-1 and Occludin proteins were dramatically augmented in RM-treated groups to maintain intestinal barrier integrity. Further analyses revealed that RM might prevent NEC pups by improving some pivotal proteins involved in oxidative stress, inflammation, and apoptosis of enterocytes, namely, by down-regulating the levels of TLR4, p-NF-κB, iNOS, cleaved Caspase-3, and Bax and up-regulating the levels of Bcl-2, NQO1, and Nrf2. CONCLUSIONS The RM prevented the intestinal inflammation and damage caused by NEC by regulating the expression of several pivotal proteins involved in oxidative stress, inflammation, and apoptosis. This study might provide a scientific basis for R. chinensis fruits as a traditional herbal medicine to prevent and/or alleviate NEC.
Collapse
Affiliation(s)
- Yi Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Ou Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, People's Republic of China
| | - Hongying Mi
- The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan Province, People's Republic of China
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China.
| |
Collapse
|
18
|
Fermented Myriophyllum aquaticum and Lactobacillus plantarum Affect the Distribution of Intestinal Microbial Communities and Metabolic Profile in Mice. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8050210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This research explores the effects of fermented Myriophyllum aquaticum (F) and Lactobacillus plantarum BW2013 (G) as new feed additives on the gut microbiota composition and metabolic profile of mice. Crude protein (p = 0.045), lipid (p = 0.000), and ash (p = 0.006) contents in Myriophyllum aquaticum (N) were improved, whereas raw fiber (p = 0.031) content was decreased after solid-state fermentation by G. Mice were fed with no additive control (CK), 10%N (N), 10%N + G (NG), 10%F (F), and 10%F + G (FG). High-throughput sequencing results showed that, compared with the CK group, Parabacteroides goldsteinii was increased in treatment groups and that Lactobacillus delbrueckii, Bacteroides vulgatus, and Bacteroides coprocola were increased in the F and FG groups. Bacteroides vulgatus and Bacteroides coprocola were increased in the F group compared with the N group. Metabolomic results showed that vitamin A, myricetin, gallic acid, and luteolin were increased in the F group compared with the N group. Reduction in LPG 18:1 concentration in the N and F groups could be attenuated or even abolished by supplementation with G. Furthermore, 9-oxo-ODA was upregulated in the FG group compared with the F group. Collectively, N, F, and G have beneficial effects on gut microbiota and metabolic profile in mice, especially intake of FG.
Collapse
|
19
|
Gürbüz M, Aktaç Ş. Understanding the role of vitamin A and its precursors in the immune system. NUTR CLIN METAB 2022. [DOI: 10.1016/j.nupar.2021.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Micronutrient Improvement of Epithelial Barrier Function in Various Disease States: A Case for Adjuvant Therapy. Int J Mol Sci 2022; 23:ijms23062995. [PMID: 35328419 PMCID: PMC8951934 DOI: 10.3390/ijms23062995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
The published literature makes a very strong case that a wide range of disease morbidity associates with and may in part be due to epithelial barrier leak. An equally large body of published literature substantiates that a diverse group of micronutrients can reduce barrier leak across a wide array of epithelial tissue types, stemming from both cell culture as well as animal and human tissue models. Conversely, micronutrient deficiencies can exacerbate both barrier leak and morbidity. Focusing on zinc, Vitamin A and Vitamin D, this review shows that at concentrations above RDA levels but well below toxicity limits, these micronutrients can induce cell- and tissue-specific molecular-level changes in tight junctional complexes (and by other mechanisms) that reduce barrier leak. An opportunity now exists in critical care—but also medical prophylactic and therapeutic care in general—to consider implementation of select micronutrients at elevated dosages as adjuvant therapeutics in a variety of disease management. This consideration is particularly pointed amidst the COVID-19 pandemic.
Collapse
|
21
|
Lu L, Xu W, Liu J, Chen L, Hu S, Sheng Q, Zhang M, Lv Z. DRG1 Maintains Intestinal Epithelial Cell Junctions and Barrier Function by Regulating RAC1 Activity in Necrotizing Enterocolitis. Dig Dis Sci 2021; 66:4237-4250. [PMID: 33471252 DOI: 10.1007/s10620-020-06812-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/29/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND An immature intestine is a high-risk factor for necrotizing enterocolitis (NEC), which is a serious intestinal disease in newborns. The regulation of developmentally regulated GTP-binding protein 1 (DRG1) during organ development suggests a potential role of DRG1 in the maturation process of the intestine. AIM To illustrate the function of DRG1 during the pathogenesis of NEC. METHODS DRG1 expression in the intestine was measured using immunohistochemistry and q-PCR. Immunoprecipitation coupled with mass spectrometry was used to identify the interacting proteins of DRG1. The biological functions of the potential interactors were annotated with the Database for Annotation, Visualization and Integrated Discovery. Caco2 and FHs74Int cells with stable DRG1 silencing or overexpression were used to investigate the influence of DRG1 on cell junctions and intestinal barrier permeability and to elucidate the downstream mechanism. RESULTS DRG1 was constitutively expressed during the intestinal maturation process but significantly decreased in the ileum in the context of NEC. Protein interaction analysis revealed that DRG1 was closely correlated with cell junctions. DRG1 deficiency destabilized the E-cadherin and occludin proteins near the cell membrane and increased the permeability of the epithelial cell monolayer, while DRG1 overexpression prevented lipopolysaccharide-induced disruption of E-cadherin and occludin expression and cell monolayer integrity. Further investigation suggested that DRG1 maintained cell junctions, especially adherens junctions, by regulating RAC1 activity, and RAC1 inhibition with NSC23766 attenuated intestinal injury and led to improved barrier integrity in experimental NEC. CONCLUSIONS Our findings illustrate the mechanism underlying the effect of DRG1 deficiency on epithelial cell permeability regulation and provide evidence supporting the application of RAC1 inhibitors for protection against NEC.
Collapse
Affiliation(s)
- Li Lu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China
| | - Weijue Xu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China
| | - Jiangbin Liu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China
| | - Liping Chen
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China
| | - Shaohua Hu
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China
| | - Qingfeng Sheng
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China
| | - Minghua Zhang
- Clinical Pharmacy Laboratory, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200040, China.
| |
Collapse
|
22
|
Hong W, Xu D, Song X, Niu B, Zhuang Z, Lu Y, Lei X, Ma R, Lu C, Sun N, Mao Y, Li X. Vitamin A and retinoic acid accelerate the attenuation of intestinal adaptability upon feeding induced by high-fat diet in mice. J Nutr Biochem 2021; 97:108803. [PMID: 34147602 DOI: 10.1016/j.jnutbio.2021.108803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/29/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
With its unique cellular plasticity, the small intestinal mucosa exhibits efficient adaptability upon feeding. However, little is known about the effect of high-fat diet (HFD) feeding on this adaption and its underlying mechanism. Herein, we demonstrated that the cell proliferation ability, mitochondrial morphology, and global transcriptomic profile of the small intestine exhibited a prominent discrepancy between the fasted and refed state in mice, which were markedly attenuated by long-term HFD feeding. The retinol (Vitamin A, VA) metabolism pathway was dramatically affected by HFD feeding in the small intestine. Both VA and its active metabolite retinoic acid (RA), with the administration of lipid micelles, promoted the expression of genes involved in lipid absorption and suppressed the expression of genes involved in the cell proliferation of intestinal organoids. Via chip-qPCR and RT-qPCR, genes involved in lipid metabolism and cell proliferation were target genes of RARα/RXRα in small intestinal organoids treated with RA and lipid micelles. The role of VA in the in vivo attenuation of intestinal adaptability, in response to HFD, was evaluated. Mice were fed a normal chow diet, HFD, or HFD diet supplemented with additional 1.5-fold VA for 12 weeks. VA supplementation in HFD accelerated the attenuation of intestinal adaptability upon feeding induced by HFD, promoted lipid absorption gene expression, and increased body weight and serum cholesterol levels. In conclusion, the discrepancy of the small intestine between the fasted and refed state was dramatically attenuated by HFD feeding, in which VA and RA might play important roles.
Collapse
Affiliation(s)
- Wenting Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dongke Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaowei Song
- Department of Chemistry, Fudan University, Shanghai, China
| | - Baolin Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziyan Zhuang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiteng Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaohong Lei
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Runjun Ma
- Center for Gastrointestinal Endoscopy, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yimin Mao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China;.
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Ji YC, Sun Q, Fu CY, She X, Liu XC, He Y, Ai Q, Li LQ, Wang ZL. Exogenous Autoinducer-2 Rescues Intestinal Dysbiosis and Intestinal Inflammation in a Neonatal Mouse Necrotizing Enterocolitis Model. Front Cell Infect Microbiol 2021; 11:694395. [PMID: 34422680 PMCID: PMC8375469 DOI: 10.3389/fcimb.2021.694395] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/21/2021] [Indexed: 12/26/2022] Open
Abstract
Autoinducer-2 (AI-2) is believed to be a bacterial interspecies signaling molecule that plays an important role in the regulation of the physiological behaviors of bacteria. The effect of AI-2 on the process of necrotizing enterocolitis (NEC) is unknown, and the aim of this study was to study the effect of AI-2 in a mouse NEC model. C57BL/6 mouse pups were randomly divided into three groups: the control group, the NEC group, and the NEC+AI-2 (NA) group. Exogenous AI-2 (500 nM) was added to the formula milk of the NA group. The concentrations of fecal AI-2 and flora were tested. The expression of cytokines, TLR4 and NF-κB in intestinal tissue was detected. The AI-2 level was significantly decreased in the NEC group (P<0.05). Compared with the NEC group, the intestinal injury scores, expression of TLR4, NF-kB, and proinflammatory factors (IL-1β, IL-6, IL-8 and TNF-α) were reduced, and expression of anti-inflammatory factor (IL-10) was increased in the NA group mice (P<0.05). At the phylum level, the Proteobacteria abundance in the NA group was significantly increased, while the Bacteroidota abundance in the control group was significantly increased (P<0.05). At the genus level, Helicobacter and Clostridium_sensu_stricto_1 exhibited significantly greater abundance in the NEC group than in the other two groups, while Lactobacillus had the opposite trend (P<0.05). In addition, the abundances of Klebsiella, Rodentibacter and Enterococcus were significantly higher in the NA group than in the NEC and control groups (P < 0.05). Exogenous AI-2 partially reverses flora disorder and decreases inflammation in an NEC mouse model.
Collapse
Affiliation(s)
- Yan-Chun Ji
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qian Sun
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chun-Yan Fu
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiang She
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiao-Chen Liu
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qing Ai
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zheng-Li Wang
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
24
|
Gomes CDC, Passos TS, Morais AHA. Vitamin A Status Improvement in Obesity: Findings and Perspectives Using Encapsulation Techniques. Nutrients 2021; 13:nu13061921. [PMID: 34204998 PMCID: PMC8228342 DOI: 10.3390/nu13061921] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 12/11/2022] Open
Abstract
The association between obesity and vitamin A has been studied. Some studies point to the anti-obesity activity related to this vitamin, carotenoids with provitamin A activity, and carotenoid conversion products. This performance has been evaluated in respect of adipogenesis, metabolic activity, oxidation processes, secretory function, and oxidative stress modulation, showing a new property attributed to vitamin A in preventing and treating obesity. However, vitamin A and its precursors are highly sensitive and easily degraded when subjected to heat, the presence of light, and oxygen, in addition to losses related to the processes of digestion and absorption. In this context, encapsulation presents itself as an alternative capable of increasing vitamin A’s stability in the face of unfavorable conditions in the environment, which can reduce its functionality. Considering that vitamin A’s status shows a strong correlation with obesity and is an innovative theme, this article addresses the associations between vitamin A’s consumption and its precursors, encapsulated or not, and its physiological effects on obesity. The present narrative review points out those recent studies that demonstrate that vitamin A and its encapsulated precursors have the most preserved functionality, which guarantees better effects on obesity therapy.
Collapse
Affiliation(s)
- Camila de Carvalho Gomes
- Postgraduate Program in Biochemistry and Molecular Biology, Center for Biosciences, Federal University of Rio Grande do Norte, Natal 59078 970, Brazil;
| | - Thais Souza Passos
- Department of Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078 970, Brazil;
| | - Ana Heloneida Araújo Morais
- Postgraduate Program in Biochemistry and Molecular Biology, Center for Biosciences, Federal University of Rio Grande do Norte, Natal 59078 970, Brazil;
- Department of Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078 970, Brazil;
- Postgraduate Program in Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078 970, Brazil
- Correspondence: ; Tel.: +55-(84)991061887
| |
Collapse
|
25
|
Zhang M, Xu Y, Zhang J, Sun Z, Ban Y, Wang B, Hou X, Cai Y, Li J, Wang M, Wang W. Application of methane and hydrogen-based breath test in the study of gestational diabetes mellitus and intestinal microbes. Diabetes Res Clin Pract 2021; 176:108818. [PMID: 33932493 DOI: 10.1016/j.diabres.2021.108818] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/07/2021] [Accepted: 04/12/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Gestational diabetes mellitus (GDM) is a common complication of pregnancy. The purpose of this study was to compare the incidence of small intestinal bacterial overgrowth (SIBO) in patients with GDM and the control group by methane and hydrogen lactulose breath test (LBT), and to explore its relationship with inflammation, vitamins, and the outcomes of maternal and child. METHODS LBT was detected in 220 GDM patients, 160 pregnancy control patients and 160 pre-pregnancy control patients. The fasting blood glucose, white blood cells, vitamin A, D, E, neonatal weight, neonatal blood glucose and so on were compared and analyzed. RESULTS There was no statistical significance in the general data of the three groups. The proportion of abdominal distension in the GDM group was higher than that in the other two groups (P < 0.001). The positive rates of SIBO + in GDM group, gestational control group and pre-pregnancy control group were 54.55%, 27.50% and 14.38%, respectively. The average abundance of hydrogen and methane in GDM group was significantly higher than that in control group at each time point. In the GDM group, SIBO + subjects had higher levels of fasting blood glucose, glycoglycated hemoglobin, C-reactive protein, neonatal weight, and lower levels of vitamin D and neonatal blood glucose (P < 0.001). CONCLUSION Patients with GDM have a high incidence of SIBO, and SIBO may further increase their blood glucose by affecting inflammatory response and vitamin level, and even affect the outcome of mother and child.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yajuan Xu
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Jingzhe Zhang
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zongzong Sun
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yanjie Ban
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Biao Wang
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaofeng Hou
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yanjun Cai
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingjing Li
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Mengqi Wang
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wentao Wang
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
26
|
de Lange IH, van Gorp C, Eeftinck Schattenkerk LD, van Gemert WG, Derikx JPM, Wolfs TGAM. Enteral Feeding Interventions in the Prevention of Necrotizing Enterocolitis: A Systematic Review of Experimental and Clinical Studies. Nutrients 2021; 13:1726. [PMID: 34069699 PMCID: PMC8161173 DOI: 10.3390/nu13051726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022] Open
Abstract
Necrotizing enterocolitis (NEC), which is characterized by severe intestinal inflammation and in advanced stages necrosis, is a gastrointestinal emergency in the neonate with high mortality and morbidity. Despite advancing medical care, effective prevention strategies remain sparse. Factors contributing to the complex pathogenesis of NEC include immaturity of the intestinal immune defense, barrier function, motility and local circulatory regulation and abnormal microbial colonization. Interestingly, enteral feeding is regarded as an important modifiable factor influencing NEC pathogenesis. Moreover, breast milk, which forms the currently most effective prevention strategy, contains many bioactive components that are known to support neonatal immune development and promote healthy gut colonization. This systematic review describes the effect of different enteral feeding interventions on the prevention of NEC incidence and severity and the effect on pathophysiological mechanisms of NEC, in both experimental NEC models and clinical NEC. Besides, pathophysiological mechanisms involved in human NEC development are briefly described to give context for the findings of altered pathophysiological mechanisms of NEC by enteral feeding interventions.
Collapse
Affiliation(s)
- Ilse H. de Lange
- European Surgical Center Aachen/Maastricht, Department of Pediatric Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), 6202 AZ Maastricht, The Netherlands; (I.H.d.L.); (W.G.v.G.)
- Department of Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | - Charlotte van Gorp
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | - Laurens D. Eeftinck Schattenkerk
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.D.E.S.); (J.P.M.D.)
| | - Wim G. van Gemert
- European Surgical Center Aachen/Maastricht, Department of Pediatric Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), 6202 AZ Maastricht, The Netherlands; (I.H.d.L.); (W.G.v.G.)
- Department of Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.D.E.S.); (J.P.M.D.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
27
|
Feng D, Chen B, Zeng B, Xiao L, Yan J, Yang T, Zhu J, Li T, Wang L, Wei H, Chen J. Fecal microbiota from children with vitamin A deficiency impair colonic barrier function in germ-free mice: The possible role of alterative bile acid metabolites. Nutrition 2021; 90:111274. [PMID: 34004414 DOI: 10.1016/j.nut.2021.111274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVE This study explores the effects of fecal microbiota from children with vitamin A (VA) deficiency on colonic mucosal barrier function. METHODS The composition of gut microbes was identified in children with different VA levels, then feces from children with normal VA or VA deficiency was collected separately and transplanted into germ-free (GF) mice, respectively. Three weeks after transplantation, the colon morphology, colonic tight junction proteins, gut microbes, and metabolites were evaluated. RESULTS In children, Bifidobacterium and Bacteroides were positively correlated with VA levels. Colonization of VA deficiency fecal microbiota markedly impaired colonic development in GF mice, down-regulated colonic tight junction-related proteins occludin and claudin-1, and reduced immunoglobulin A secretion. Furthermore, fecal microbiota transplantation with different VA levels altered composition of gut microbes and bile acid metabolism pathways in GF mice. CONCLUSION These data suggest that fecal microbiota from children with VA deficiency attenuates colonic barrier function in GF mice, which may be achieved by changing the bile acid metabolic pathways.
Collapse
Affiliation(s)
- Di Feng
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Baolin Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Benhua Zeng
- Laboratory Animal Department, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Lu Xiao
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Junyan Yan
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Ting Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Jiang Zhu
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Lan Wang
- Shanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Taiyuan, China
| | - Hong Wei
- Laboratory Animal Department, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University; Chongqing Key Laboratory of Childhood Nutrition and Health; Ministry of Education Key Laboratory of Child Development and disorders; National Clinical Research Center for Child Health and Disorders, Chongqing, China.
| |
Collapse
|
28
|
Ran X, He Y, Ai Q, Shi Y. Effect of antibiotic-induced intestinal dysbacteriosis on bronchopulmonary dysplasia and related mechanisms. J Transl Med 2021; 19:155. [PMID: 33874953 PMCID: PMC8054697 DOI: 10.1186/s12967-021-02794-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/16/2021] [Indexed: 12/30/2022] Open
Abstract
Background Modification of the gut microbiota by antibiotics may influence the disease susceptibility and immunological responses. Infants in the neonatal intensive care unit (NICU) subjected to frequent antibiotics and oxygen therapies, which may give rise to local and systemic inflammatory reactions and progression of bronchopulmonary dysplasia (BPD). This study aimed to investigate the role of intestinal dysbacteriosis by antibiotic therapy before hyperoxia exposure in the progression of BPD. Methods Mice had been exposed to hyperoxia (85% O2) since postnatal day 3 until day 16 for the BPD model establishment, treated with antibiotics from postnatal day 2 until day 8. Treated mice and appropriate controls were harvested on postnatal day 2 or 10 for 16S rRNA gene sequencing, or postnatal day 17 for assessment of alveolar morphometry and macrophages differentiation. Results Antibiotic-induced intestinal dysbacteriosis before hyperoxia exposure gave rise to deterioration of BPD evidenced by reduced survival rates and alveolarization. Moreover, antibiotic-induced intestinal dysbacteriosis resulted in increased M1 macrophage maker (iNOS) and decreased M2 macrophage maker (Arg-1) levels in lung homogenates. Conclusion Broad-spectrum antibiotic-induced intestinal dysbacteriosis may participate in BPD pathogenesis via alteration of the macrophage polarization status. Manipulating the gut microbiota may potentially intervene the therapy of BPD. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02794-6.
Collapse
Affiliation(s)
- Xiao Ran
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders, No.136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders, No.136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qing Ai
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders, No.136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders, No.136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, People's Republic of China. .,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China. .,Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
29
|
Relationship between Nutrient Intake and Human Gut Microbiota in Monozygotic Twins. ACTA ACUST UNITED AC 2021; 57:medicina57030275. [PMID: 33809761 PMCID: PMC8002349 DOI: 10.3390/medicina57030275] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Background and Objectives: The gut microbiota is associated with human health and dietary nutrition. Various studies have been reported in this regard, but it is difficult to clearly analyze human gut microbiota as individual differences are significant. The causes of these individual differences in intestinal microflora are genetic and/or environmental. In this study, we focused on differences between identical twins in Japan to clarify the effects of nutrients consumed on the entire gut microbiome, while excluding genetic differences. Materials and Methods: We selected healthy Japanese monozygotic twins for the study and confirmed their zygosity by matching 15 short tandem repeat loci. Their fecal samples were subjected to 16S rRNA sequencing and bioinformatics analyses to identify and compare the fluctuations in intestinal bacteria. Results: We identified 12 genera sensitive to environmental factors, and found that Lactobacillus was relatively unaffected by environmental factors. Moreover, we identified protein, fat, and some nutrient intake that can affect 12 genera, which have been identified to be more sensitive to environmental factors. Among the 12 genera, Bacteroides had a positive correlation with retinol equivalent intake (rs = 0.38), Lachnospira had a significantly negative correlation with protein, sodium, iron, vitamin D, vitamin B6, and vitamin B12 intake (rs = −0.38, −0.41, −0.39, −0.63, −0.42, −0.49, respectively), Lachnospiraceae ND3007 group had a positive correlation with fat intake (rs = 0.39), and Lachnospiraceae UCG-008 group had a negative correlation with the saturated fatty acid intake (rs = −0.45). Conclusions: Our study is the first to focus on the relationship between human gut microbiota and nutrient intake using samples from Japanese twins to exclude the effects of genetic factors. These findings will broaden our understanding of the more intuitive relationship between nutrient intake and the gut microbiota and can be a useful basis for finding useful biomarkers that contribute to human health.
Collapse
|
30
|
The relationship between umbilical cord blood vitamin A levels and late preterm infant morbidities: a prospective cohort study. Eur J Pediatr 2021; 180:791-797. [PMID: 32851492 DOI: 10.1007/s00431-020-03787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
The aim of this study is to explore the association between umbilical cord blood (UCB) vitamin A levels and late preterm infants morbidities. We conducted a prospective cohort study of 208 late-preterm infants(from 34 0/7 to 36 6/7 weeks gestational age) between January 1, 2014 and June 30, 2015. UCB specimens were collected shortly after birth, and vitamin A levels were determined by enzyme-linked immunosorbent assay. Prevalence of low UCB vitamin A level < 0.7 μmol/L was 37.5% in late preterm infants. In comparison to vaginal delivery, cesarean section was associated with UCB vitamin A level < 0.7 μmol/L (P < 0.001). Nevertheless, UCB vitamin A levels did not correlate with gestational age, birth weight, and gender. UCB vitamin A level < 0.7 μmol/L was not an independent risk factor for hospitalization, oxygen supplementation, hyperbilirubinemia, sepsis, and respiratory distress syndrome.Conclusions: Low umbilical cord blood vitamin A levels are common among late-preterm infants. Cesarean section delivery is associated with low umbilical cord blood vitamin A level. Low umbilical cord blood vitamin A levels at birth do not increase morbidity of late-preterm infants, including hyperbilirubinemia, sepsis, and respiratory distress syndrome. What is Known: • Late preterm infants have a higher morbidity and mortality rates when compared to term infants. • Low plasma vitamin A levels increase the risk of preterm infants' morbidity. What is New: • Late preterm infants commonly have low level of umbilical cord blood vitamin A. • Low umbilical cord blood vitamin A level at birth appears to be not associated with the morbidity of late-preterm infants. • Cesarean section is associated with low umbilical cord blood vitamin A level < 0.7 μmol/L compared with vaginal delivery.
Collapse
|
31
|
Griffiths V, Al Assaf N, Khan R. Review of claudin proteins as potential biomarkers for necrotizing enterocolitis. Ir J Med Sci 2021; 190:1465-1472. [PMID: 33492576 PMCID: PMC8521514 DOI: 10.1007/s11845-020-02490-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
Background Claudin proteins are a component of tight junctions found in cell-cell adhesion complexes. A central feature of necrotizing enterocolitis (NEC) is intestinal permeability, with changes to claudin proteins potentially contributing to intestinal instability, inflammation, and the progression of NEC. A current area of interest is the development of a novel, noninvasive biomarker for the detection of NEC in neonates at risk of developing this disease, in order to reduce morbidity and mortality through earlier intervention. Aims This review aims to explore the relevance of claudin proteins in the pathophysiology of NEC and their potential usefulness as a biomarker. Methods This review was conducted using the search terms “claudin” + “necrotizing enterocolitis”, with 27 papers selected for review. Results Claudin proteins appear to have a role in the stability of the gut epithelium through the regulation of intestinal permeability, maturity, and inflammation. Formula feeding has been shown to promote inflammation and result in changes to claudin proteins, while breastfeeding and certain nutritional supplements lead to reduced inflammation and improved intestinal stability as demonstrated through changes to claudin protein expression. Preliminary studies in human neonates suggest that urinary claudin measurements may be used to predict the development of NEC. Conclusions Alterations to claudin proteins may reflect changes seen to intestinal permeability and inflammation in the context of NEC. Further research is necessary to understand the relevance of claudin proteins in the pathophysiology of NEC and their use as a biomarker.
Collapse
Affiliation(s)
- Victoria Griffiths
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland.
| | - Niazy Al Assaf
- Department of Neonatology, University Maternity Hospital Limerick, Limerick, Ireland
| | - Rizwan Khan
- Department of Neonatology, University Maternity Hospital Limerick, Limerick, Ireland
| |
Collapse
|
32
|
Abstract
Mucosal surfaces are distinctive sites exposed to environmental, dietary, and microbial antigens. Particularly in the gut, the host continuously actively adapts via complex interactions between the microbiota and dietary compounds and immune and other tissue cells. Regulatory T cells (Tregs) are critical for tuning the intestinal immune response to self- and non-self-antigens in the intestine. Its importance in intestinal homeostasis is illustrated by the onset of overt inflammation caused by deficiency in Treg generation, function, or stability in the gut. A substantial imbalance in Tregs has been observed in intestinal tissue during pathogenic conditions, when a tightly regulated and equilibrated system becomes dysregulated and leads to unimpeded and chronic immune responses. In this chapter, we compile and critically discuss the current knowledge on the key factors that promote Treg-mediated tolerance in the gut, such as those involved in intestinal Treg differentiation, specificity and suppressive function, and their immunophenotype during health and disease. We also discuss the current state of knowledge on Treg dysregulation in human intestine during pathological states such as inflammatory bowel disease (IBD), necrotizing enterocolitis (NEC), graft-versus-host disease (GVHD), and colorectal cancer (CRC), and how that knowledge is guiding development of Treg-targeted therapies to treat or prevent intestinal disorders.
Collapse
|
33
|
The microbiota protects against Pseudomonas aeruginosa pneumonia via γδ T cell-neutrophil axis in mice. Microbes Infect 2020; 22:294-302. [DOI: 10.1016/j.micinf.2020.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
|
34
|
Suzuki T. Regulation of the intestinal barrier by nutrients: The role of tight junctions. Anim Sci J 2020; 91:e13357. [PMID: 32219956 PMCID: PMC7187240 DOI: 10.1111/asj.13357] [Citation(s) in RCA: 322] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022]
Abstract
Tight junctions (TJs) play an important role in intestinal barrier function. TJs in intestinal epithelial cells are composed of different junctional molecules, such as claudin and occludin, and regulate the paracellular permeability of water, ions, and macromolecules in adjacent cells. One of the most important roles of the TJ structure is to provide a physical barrier to luminal inflammatory molecules. Impaired integrity and structure of the TJ barrier result in a forcible activation of immune cells and chronic inflammation in different tissues. According to recent studies, the intestinal TJ barrier could be regulated, as a potential target, by dietary factors to prevent and reduce different inflammatory disorders, although the precise mechanisms underlying the dietary regulation remain unclear. This review summarizes currently available information on the regulation of the intestinal TJ barrier by food components.
Collapse
Affiliation(s)
- Takuya Suzuki
- Department of Biofunctional Science and Technology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Japan.,Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
35
|
All-trans retinoic acid reduces the transcriptional regulation of intestinal sodium-dependent phosphate co-transporter gene (Npt2b). Biochem J 2020; 477:817-831. [PMID: 32016357 DOI: 10.1042/bcj20190716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/13/2020] [Accepted: 02/03/2020] [Indexed: 01/03/2023]
Abstract
Inorganic phosphate (Pi) homeostasis is regulated by intestinal absorption via type II sodium-dependent co-transporter (Npt2b) and by renal reabsorption via Npt2a and Npt2c. Although we previously reported that vitamin A-deficient (VAD) rats had increased urine Pi excretion through the decreased renal expression of Npt2a and Npt2c, the effect of vitamin A on the intestinal Npt2b expression remains unclear. In this study, we investigated the effects of treatment with all-trans retinoic acid (ATRA), a metabolite of vitamin A, on the Pi absorption and the Npt2b expression in the intestine of VAD rats, as well as and the underlying molecular mechanisms. In VAD rats, the intestinal Pi uptake activity and the expression of Npt2b were increased, but were reduced by the administration of ATRA. The transcriptional activity of reporter plasmid containing the promoter region of the rat Npt2b gene was reduced by ATRA in NIH3T3 cells overexpressing retinoic acid receptor (RAR) and retinoid X receptor (RXR). On the other hand, CCAAT/enhancer-binding proteins (C/EBP) induced transcriptional activity of the Npt2b gene. Knockdown of the C/EBP gene and a mutation analysis of the C/EBP responsible element in the Npt2b gene promoter indicated that C/EBP plays a pivotal role in the regulation of Npt2b gene transcriptional activity by ATRA. EMSA revealed that the RAR/RXR complex inhibits binding of C/EBP to Npt2b gene promoter. Together, these results suggest that ATRA may reduce the intestinal Pi uptake by preventing C/EBP activation of the intestinal Npt2b gene.
Collapse
|
36
|
Fu CY, Li LQ, Yang T, She X, Ai Q, Wang ZL. Autoinducer-2 May Be a New Biomarker for Monitoring Neonatal Necrotizing Enterocolitis. Front Cell Infect Microbiol 2020; 10:140. [PMID: 32373545 PMCID: PMC7179697 DOI: 10.3389/fcimb.2020.00140] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/18/2020] [Indexed: 12/19/2022] Open
Abstract
Autoinducer-2 (AI-2) has a widely accepted role in bacterial intra- and interspecies communication. Little is known about the relationships between AI-2 and NEC. This study found that AI-2 levels in patients and in a NEC mouse model were detected using the Vibrio harveyi BB170 assay system. Bacterial communities of the newborns' stool microbiota (NEC acute group, NEC recovery group, control group, and antibiotics-free group) and of the NEC mouse model (NEC group and control group) were detected by high-throughput sequencing. Intestinal histopathological changes were observed after HE staining. The AI-2 level in the NEC acute group (44.75 [40.17~65.52]) was significantly lower than that in the control group, NEC recovery group and antibiotics-free group. The overall microbiota compositions of each group at the phylum level were not significantly different. The proportions of Enterococcus, Clostridium_sensu_stricto_1, Peptoclostridium, and Veillonella had significant differences among the 4 groups at the genus level. In animal experiments, the AI-2 level in feces of NEC mice (56.89 ± 11.87) was significantly lower than that in the feces of control group mice (102.70 ± 22.97). The microbiota compositions of NEC and control group mice at the phylum level were not significantly different. At the genus level, Klebsiella, Clostridium_sensu_stricto_1, and Peptoclostridium abundances in the NEC group increased significantly compared with those in the control group (P < 0.05). In addition, Lactobacillus, Pasteurella, and Parabacteroides abundances in the NEC group decreased significantly compared with those in the normal control group (P < 0.05), while Lactobacillus, Pasteurella, and Parabacteroides abundances had the opposite trend. The AI-2 concentration decreased significantly in the acute phase of NEC and increased gradually in the convalescent phase. We conclude that the concentration of AI-2 was correlated with intestinal flora disorder and different stages of disease. AI-2 may be a new biomarker for the diagnosis and monitoring of NEC. Trial Registry: ClinicalTrials.gov; ChiCTR-ROC-17013746; URL: www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Chun-Yan Fu
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Lu-Quan Li
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Ting Yang
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Xiang She
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Qing Ai
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Zheng-Li Wang
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| |
Collapse
|
37
|
Wang YG, Gao Y, Feng J, Dou YQ. Effect of Modified Xijiao Dihuang Decoction () on Intestinal Flora and Th17/Treg in Rats with Radiation Enteritis. Chin J Integr Med 2020; 27:198-205. [PMID: 32240475 DOI: 10.1007/s11655-020-3261-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To observe the effect of Modified Xijiao Dihuang Decoction (, MXDD) on rats with radiation enteritis, and explore its action mechanism. METHODS Thirty female Sprague Dawley rats were divided into the control, model, dexamethasone (DXM), golden bifid (GB) and MXDD groups using random number table, 6 rats in each group. Except the control group, the other rats were developed into radiation enteritis model by exposing to a single 60Co-γ ray at a dose of 11 Gy. The rats in the DXM, GB and MXDD groups were treated with DXM (1.425 mg/kg), GB (0.8 g/kg) and MXDD (36.0 g/kg) for 3 days, respectively. Body weight and diarrhea condition of rats were evaluated daily. On day 3, the feces of rats were collected for intestinal flora detection and the small intestinal tissues were also collected. Bacterial species annotation, alpha and beta diversities as well as composition of intestinal flora were detected and compared. The protein and mRNA expressions of interleukin 17 (IL-17), retinoid-related orphan nuclear receptor gamma t (ROR-γt) and forkhead/ winged helix transcription factor p3 (FoxP3) were determined by Western blot and polymerase chain reaction, respectively. The abundance and diversity of intestinal flora as well as the proportion at the phylum and genus levels were assayed by 16S rRNA metagenome sequencing. Correlation between intestinal flora and Th17/Treg was analyzed by heatmap method. RESULTS On day 1 to 3 after radiation, compared with the control group, the body weight in model group was decreased (P<0.05 or P<0.01). Compared with the model group, MXDD could alleviate weight loss and diarrhea caused by irradiation. At the phylum level, MXDD cause a significant increase in Firmicutes, and a decrease in Proteobacteria (P<0.05 or P<0.01). At the genus level, MXDD reduced the proportion of Escherichia Shigella (P<0.01). In addition, IL-17 and FoxP3 mRNA and protein expression levels were down-regulated and ROR-γt was up-regulated by MXDD treatment (P<0.05). Besides, Firmicutes and Lactobacillus were positively correlated with FoxP3 (r=0.73, 0.79, respectively; P<0.01), negatively correlated with IL-17 (r=0.66, 0.64, respectively; P<0.01 or P<0.05) and ROR-γt (r0.73, 0.81, respectively; P<0.01). Proteobacteria and Escherichia Shigella both had positive correlation with IL-17 (r 0.77, 0.57, respectively; P<0.01 or P<0.05 ) and ROR-γt (r=0.94, 0.79, respectively; P<0.01) and negative correlation with FoxP3 (r0.74, 0.65; P<0.01). CONCLUSION MXDD could improve the survival status of irradiated rats by regulating the richness, diversity and composition of intestinal flora, and restoring the balance of Th17/Treg.
Collapse
Affiliation(s)
- Yu-Guo Wang
- Medical School of Chinese People's Liberation Army, The General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Yue Gao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jian Feng
- Medical School of Chinese People's Liberation Army, The General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Yong-Qi Dou
- Medical School of Chinese People's Liberation Army, The General Hospital of People's Liberation Army, Beijing, 100853, China.
| |
Collapse
|
38
|
Liang S, Lai P, Li X, Xu J, Bao Y, Fang Y, Ding M. Ulinastatin Reduces the Severity of Intestinal Damage in the Neonatal Rat Model of Necrotizing Enterocolitis. Med Sci Monit 2019; 25:9123-9130. [PMID: 31786582 PMCID: PMC6904988 DOI: 10.12659/msm.919413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Ulinastatin is a protease inhibitor derived from urine that has shown anti-inflammatory effects in human disease, including in sepsis. Necrotizing enterocolitis (NEC) is a common gastrointestinal disease in premature infants. Our aim was to explore the effects of ulinastatin on a neonatal NEC rat model. Material/Methods Forty-five neonatal rats were divided into 3 groups: normal control; NEC+sepsis-induced kidney injury (SIRS); NEC/SIRS+ulinastatin. The NEC/SIRS model was induced by injection of intraperitoneal saline, enteral formula feeding, hypoxia-hyperoxide, and cold stress exposure. The NEC/SIRS neonatal rats were perfused with ulinastatin at a dose of 10 000 u/kg/day. Giemsa staining and hematoxylin and eosin (H&E) were performed to evaluate the severity of intestinal damage. To assess intestinal cell apoptosis, we examined the expression of caspase-3 by TUNEL staining and western blot analysis. Intestinal levels of inflammatory cytokines (IL-1β, IL-6, and TNF-α) were examined using ELISA assay. Results Rats in the NEC treated with ulinastatin group had better physiological status and histological score compared to the NEC/SIRS group. Ulinastatin reduced NEC-induced weight loss. Macroscopic and microscopic morphology analyses showed that rats in the NEC treated with ulinastatin group had lower severity of intestinal damage compared to the NEC/SIRS group. TUNEL staining and caspase-3 expression detection results revealed that ulinastatin significantly inhibited intestinal cell apoptosis of NEC. Furthermore, ulinastatin decreased the intestinal levels of IL-1β, IL-6, and TNF-α in NEC. Conclusions Ulinastatin could ameliorate the severity of intestinal damage in NEC and possess anti-apoptosis and anti-inflammation effects.
Collapse
Affiliation(s)
- Shuxia Liang
- Department of Ophthalmology, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang, China (mainland)
| | - Panjian Lai
- Department of Pediatrics, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang, China (mainland)
| | - Xiaobing Li
- Department of Pediatrics, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang, China (mainland)
| | - Jie Xu
- Operating Room, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang, China (mainland)
| | - Yunguang Bao
- Department of Pediatrics, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang, China (mainland)
| | - Yuanshu Fang
- Department of Laboratory Animals Center, Jinhua Institute for Food and Drug Control, Jinhua, Zhejiang, China (mainland)
| | - Mingxing Ding
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang, China (mainland)
| |
Collapse
|
39
|
Recent Advances in Prevention and Therapies for Clinical or Experimental Necrotizing Enterocolitis. Dig Dis Sci 2019; 64:3078-3085. [PMID: 30989465 DOI: 10.1007/s10620-019-05618-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/08/2019] [Indexed: 01/09/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most severe diseases of preterm neonates and has a high mortality rate. With the development of inspection techniques and new biomarkers, the diagnostic accuracy of NEC is constantly improving. The most recognized potential risk factors include prematurity, formula-feeding, infection, and microbial dysbiosis. With further understanding of the pathogenesis, more effective prevention and therapies will be applied to clinical or experimental NEC. At present, such new potential prevention and therapies for NEC are mainly focused on the Toll-like receptor 4 inflammatory signaling pathway, the repair of intestinal barrier function, probiotics, antioxidative stress, breast-feeding, and immunomodulatory agents. Many new studies have changed our understanding of the pathogenesis of NEC and improve our approaches for preventing and treating of NEC each year. This review provides an overview of the recent researches focused on clinical or experimental NEC and highlights the advances made within the past 5 years toward the development of new potential preventive approaches and therapies for this disease.
Collapse
|
40
|
Yamada S, Kanda Y. Retinoic acid promotes barrier functions in human iPSC-derived intestinal epithelial monolayers. J Pharmacol Sci 2019; 140:337-344. [DOI: 10.1016/j.jphs.2019.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
|
41
|
Shen H, Lei Y, He X, Liu D, He Z. Role of lactadherin in intestinal barrier integrity in experimental neonatal necrotizing enterocolitis. J Cell Biochem 2019; 120:19509-19517. [PMID: 31265168 DOI: 10.1002/jcb.29255] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/29/2022]
Abstract
Necrotizing enterocolitis (NEC) is one of the most widespread and devastating gastrointestinal diseases in neonates. Destruction of the intestinal barrier is the main underlying cause of NEC. The aim of this study was to determine the role of lactadherin in preventing NEC in a neonatal rat model and investigate the molecular mechanism of lactadherin-mediated protection of the intestinal barrier. Neonatal rats were divided into three groups: dam feeding (DF), NEC (NEC), and NEC supplemented with 10 μg/(g·day) recombinant human lactadherin (NEC+L). Intestinal permeability, tissue damage, and cell junction protein expression and localization were evaluated. We found that lactadherin reduced weight loss caused by NEC, reduced the incidence of NEC from 100% to 46.7%, and reduced the mean histological score for tissue damage to 1.40 compared with 2.53 in the NEC group. Intestinal permeability of lactadherin-treated rats was significantly reduced when compared with that of the NEC group. In addition, the expression levels of JAM-A, claudin 3, and E-calcium in the ileum of NEC group animals increased compared with those in the ileum of DF group animals, and these levels decreased in the NEC+L group. Lactadherin changed the localization of claudin 3, occludin, and E-cadherin in epithelial cells. The mechanism underlying lactadherin-mediated protection of the intestinal barrier might be restoring the correct expression levels and localization of tight junction and adherent junction proteins. These findings suggest a new candidate agent for the prevention of NEC in newborns.
Collapse
Affiliation(s)
- Haiqing Shen
- Department of Neonatology, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Perinatal Research Laboratory, Xin-Hua Hospital, Shanghai Institute of Pediatric Research Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Lei
- Department of Neonatology, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuemei He
- Department of Neonatology, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danyang Liu
- Department of Neonatology, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Perinatal Research Laboratory, Xin-Hua Hospital, Shanghai Institute of Pediatric Research Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenjuan He
- Department of Neonatology, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Perinatal Research Laboratory, Xin-Hua Hospital, Shanghai Institute of Pediatric Research Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Xiao L, Chen B, Feng D, Yang T, Li T, Chen J. TLR4 May Be Involved in the Regulation of Colonic Mucosal Microbiota by Vitamin A. Front Microbiol 2019; 10:268. [PMID: 30873131 PMCID: PMC6401601 DOI: 10.3389/fmicb.2019.00268] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/01/2019] [Indexed: 12/14/2022] Open
Abstract
Objectives: To investigate the specific role of Toll-like receptor 4 (TLR4) in the regulation of the intestinal mucosa-associated microbiota by vitamin A (VA). Methods: Both TLR4-/- (knockout, KO) and wild-type (WT) female mice were randomly fed a VA normal (VAN) or VA deficient (VAD) diet for 4 weeks to establish the following four mouse model groups: TLR4-/- mice fed a VAN diet (KO VAN), TLR4-/- mice fed a VAD diet (KO VAD), WT mice fed a VAN diet (WT VAN), and WT mice fed a VAD diet (WT VAD). Then, the mice from each experimental group were mated with male mice with the same genetic background. The pups in the KO VAD and WT VAD groups were subsequently fed the VAD diet after weaning, while the pups in the KO VAN and WT VAN groups were fed the VAN diet continuously after weaning. The serum retinol levels of 7-week-old offspring were determined using high-performance liquid chromatography, and colons were collected from mice in each group and analyzed via 16S rRNA gene sequencing using an Illumina MiSeq platform to characterize the overall microbiota of the samples. Results: The abundance and evenness of the colon mucosa-associated microbiota were unaffected by dietary VA and TLR4 KO. VAD decreased the abundance of Anaerotruncus (Firmicutes), Oscillibacter (Firmicutes), Lachnospiraceae _NK4A136 _group (Firmicutes) and Mucispirillum (Deferribacteres) and increased the abundance of Parasutterella (Proteobacteria). TLR4 KO decreased the abundance of Bacteroides (Bacteroidetes) and Alloprevotella (Bacteroidetes). However, the abundance of Allobaculum (Firmicutes), Ruminiclostridium_9 (Firmicutes), Alistipes (Bacteroidetes), and Rikenellaceae_RC9 (Bacteroidetes) impacted the interaction between VA and TLR4. Conclusion: TLR4 may play a pivotal role in regulation of the intestinal mucosa-associated microbiota by VA to maintain the intestinal microecology.
Collapse
Affiliation(s)
- Lu Xiao
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Baolin Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Di Feng
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Ting Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| |
Collapse
|
43
|
Li B, Hock A, Wu RY, Minich A, Botts SR, Lee C, Antounians L, Miyake H, Koike Y, Chen Y, Zani A, Sherman PM, Pierro A. Bovine milk-derived exosomes enhance goblet cell activity and prevent the development of experimental necrotizing enterocolitis. PLoS One 2019; 14:e0211431. [PMID: 30699187 PMCID: PMC6353182 DOI: 10.1371/journal.pone.0211431] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/14/2019] [Indexed: 01/19/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is characterized by intestinal injury and impaired mucin synthesis. We recently showed that breast milk exosomes from rodents promote intestinal cell viability, epithelial proliferation, and stem cell activity, but whether they also affect mucus production is unknown. Therefore, the aim of this study was to investigate the effects of bovine milk-derived exosomes on goblet cell expression in experimental NEC and delineate potential underlying mechanisms of action. Exosomes were isolated from bovine milk by ultracentrifugation and confirmed by Nanoparticle Tracking Analysis and through the detection of exosome membrane markers. To study the effect on mucin production, human colonic LS174T cells were cultured and exposed to exosomes. Compared to control, exosomes promoted goblet cell expression, as demonstrated by increased mucin production and relative expression levels of goblet cell expression markers trefoil factor 3 (TFF3) and mucin 2 (MUC2). In addition, exosome treatment enhanced the expression of glucose-regulated protein 94 (GRP94), the most abundant intraluminal endoplasmic reticulum (ER) chaperone protein that aids in protein synthesis. Furthermore, experimental NEC was induced in mouse pups by hyperosmolar formula feeding, lipopolysaccharide administration and hypoxia exposure on postnatal days 5-9. Milk exosomes were given with each gavage feed. NEC was associated with ileal morphological injury and reduction in MUC2+ goblet cells and GRP94+ cells per villus. Exosome administration to NEC pups prevented these changes. This research highlights the potential novel application of milk-derived exosomes in preventing the development of NEC in high-risk infants when breast milk is not available.
Collapse
Affiliation(s)
- Bo Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Alison Hock
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Richard Y. Wu
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Adam Minich
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Steven R. Botts
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carol Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hiromu Miyake
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Yuhki Koike
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Yong Chen
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Augusto Zani
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philip M. Sherman
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Agostino Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
44
|
de Medeiros PHQS, Pinto DV, de Almeida JZ, Rêgo JMC, Rodrigues FAP, Lima AÂM, Bolick DT, Guerrant RL, Oriá RB. Modulation of Intestinal Immune and Barrier Functions by Vitamin A: Implications for Current Understanding of Malnutrition and Enteric Infections in Children. Nutrients 2018; 10:nu10091128. [PMID: 30134532 PMCID: PMC6164597 DOI: 10.3390/nu10091128] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/11/2018] [Accepted: 08/17/2018] [Indexed: 12/24/2022] Open
Abstract
The micronutrient vitamin A refers to a group of compounds with pleiotropic effects on human health. These molecules can modulate biological functions, including development, vision, and regulation of the intestinal barrier. The consequences of vitamin A deficiency and supplementation in children from developing countries have been explored for several years. These children live in an environment that is highly contaminated by enteropathogens, which can, in turn, influence vitamin A status. Vitamin A has been described to modulate gene expression, differentiation and function of diverse immune cells; however, the underlying mechanisms are not fully elucidated. This review aims to summarize the most updated advances on elucidating the vitamin A effects targeting intestinal immune and barrier functions, which may help in further understanding the burdens of malnutrition and enteric infections in children. Specifically, by covering both clinical and in vivo/in vitro data, we describe the effects of vitamin A related to gut immune tolerance/homeostasis, intestinal barrier integrity, and responses to enteropathogens in the context of the environmental enteric dysfunction. Some of the gaps in the literature that require further research are also highlighted.
Collapse
Affiliation(s)
- Pedro Henrique Q S de Medeiros
- Laboratory of Infectious Diseases, Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Daniel V Pinto
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - Juliana Zani de Almeida
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - Juliana M C Rêgo
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
- Department of Nutrition, Christus University Center, Fortaleza 60190-060 CE, Brazil.
| | - Francisco A P Rodrigues
- Laboratory of Infectious Diseases, Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - Aldo Ângelo M Lima
- Laboratory of Infectious Diseases, Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - David T Bolick
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Reinaldo B Oriá
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| |
Collapse
|