1
|
Wang X, Yin L, Geng C, Zhang J, Li J, Huang P, Li Y, Wang Q, Yang H. Impact of different feed intake levels on intestinal morphology and epithelial cell differentiation in piglets. J Anim Sci 2025; 103:skae262. [PMID: 39238159 PMCID: PMC11705090 DOI: 10.1093/jas/skae262] [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: 06/18/2024] [Accepted: 09/05/2024] [Indexed: 09/07/2024] Open
Abstract
This study aimed to investigate the effect of feed intake levels on the development of intestinal morphology and epithelial cell differentiation in piglets. Sixty-four 35-d-old healthy weaned piglets ([Large White × Landrace] × Duroc) with an initial weight (6.93 ± 0.12 kg) were randomly divided into 4 groups (100%, 80%, 40%, and 20% feed intake) with 8 replicates of 2 pigs each. Samples were collected on days 3 and 7. The results revealed that with an increase in feed restriction degree and time, the body weight and organ index of piglets significantly decreased, and the villus height (VH) and crypt depth of the duodenum, jejunum, and ileum also decreased linearly (P < 0.05). After 3 d of feed restriction, jejunal ki67, endocrine cells, goblet cells, and villus endocrine/VH all decreased linearly, but the villus cup/VH ratio increased linearly, and the 40% and 20% were significantly higher than those of the 100% and 80% (P < 0.05). There was also a linear decrease in jejunal ki67, endocrine cells, goblet cells, and villous endocrine/VH in piglets fed 7 d of food restriction; however, the villus goblet cells/VH ratio in the 20% was significantly higher than that in the 40% group and was not different from that in the 80% (P < 0.05). During 3 d of feed restriction, the expression of jejunal differentiation marker genes showed a linear decreasing trend (P < 0.05) but increased linearly after 7 d of feed restriction. The expression levels of interleukin17 (IL-17) and IL-22 also increased linearly (P < 0.05). Kyoto Encyclopedia of Genes and Genomes and gene set enrichment analysis analyses indicated that the PPAR signaling pathway, ECM-receptor interaction, and Th1, Th2, and Th17 cell differentiation were significantly enriched in these processes. real-time quantitative polymerase chain reaction demonstrated that both PPAR and ECM-receptor interactions were significantly activated during 7 d of feeding restriction (P < 0.05). The results showed that with an increase in feed restriction intensity and time, the intestinal morphology and epithelial cell proliferation and differentiation were significantly reduced, except for the goblet cells. This phenomenon is related to the regulation of intestinal differentiation by IL-17 and IL-22 secreted by the Th cells.
Collapse
Affiliation(s)
- Xin Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Lanmei Yin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Chunchun Geng
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Jiaqi Zhang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Jianzhong Li
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Pengfei Huang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yali Li
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Qiye Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Huansheng Yang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| |
Collapse
|
2
|
Perez KM, Strobel KM, Hendrixson DT, Brandon O, Hair AB, Workneh R, Abayneh M, Nangia S, Hoban R, Kolnik S, Rent S, Salas A, Ojha S, Valentine GC. Nutrition and the gut-brain axis in neonatal brain injury and development. Semin Perinatol 2024; 48:151927. [PMID: 38897828 DOI: 10.1016/j.semperi.2024.151927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Early nutritional exposures, including during embryogenesis and the immediate postnatal period, affect offspring outcomes in both the short- and long-term. Alterations of these modifiable exposures shape the developing gut microbiome, intestinal development, and even neurodevelopmental outcomes. A gut-brain axis exists, and it is intricately connected to early life feeding and nutritional exposures. Here, we seek to discuss the (1) origins of the gut-brain access and relationship with neurodevelopment, (2) components of human milk (HM) beyond nutrition and their role in the developing newborn, and (3) clinical application of nutritional practices, including fluid management and feeding on the development of the gut-brain axis, and long-term neurodevelopmental outcomes. We conclude with a discussion on future directions and unanswered questions that are critical to provide further understanding and insight into how clinicians and healthcare providers can optimize early nutritional practices to ensure children not only survive, but thrive, free of neurodevelopmental impairment.
Collapse
Affiliation(s)
- Krystle M Perez
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America
| | - Katie M Strobel
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America
| | - D Taylor Hendrixson
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America
| | - Olivia Brandon
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America
| | - Amy B Hair
- Division of Neonatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Redeat Workneh
- St Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Mahlet Abayneh
- St Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Sushma Nangia
- Department of Neonatology, Lady Hardinge Medical College and Kalawati Saran Children's Hospital, New Delhi, India
| | - Rebecca Hoban
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America
| | - Sarah Kolnik
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America
| | - Sharla Rent
- Division of Neonatology, Duke University, Durham, NC, United States of America
| | - Ariel Salas
- Department of Pediatrics, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Shalini Ojha
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Gregory C Valentine
- Division of Neonatology, University of Washington/Seattle Children's Hospital, Seattle, WA, United States of America; Department of Oral Health Sciences, University of Washington, Seattle, WA, United States of America; Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
3
|
Elefson SK, Stoll B, Davis TA, Fiorotto ML, El-Kadi SW, Genovese K, Thymann T, Sangild PT, Burrin DG. Adverse Metabolic Phenotypes in Parenterally Fed Neonatal Pigs Do Not Persist into Adolescence. J Nutr 2024; 154:638-647. [PMID: 38181968 PMCID: PMC10900187 DOI: 10.1016/j.tjnut.2023.12.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Nutrition during fetal and neonatal life is an important determinant for the risk of adult-onset diseases, especially type 2 diabetes and obesity. OBJECTIVES We aimed to determine whether total parenteral nutrition (TPN) compared with enteral formula feeding [enteral nutrition (EN)] in term piglets during the first 2 wk after birth would increase the long-term (5-mo) development of metabolic syndrome phenotypes with adverse glucose homeostasis, fatty liver disease, and obesity. METHODS Neonatal female pigs were administered TPN (n = 12) or fed enterally with a liquid enteral milk-replacer formula (EN, n = 12) for 14 d. After transitioning TPN pigs to enteral feeding of liquid formula (days 15-26), both groups were adapted to a solid high-fat diet (30% of the total diet) and sucrose (20% of the total diet) diet (days 27-33), which was fed until the end of the study (140 d). Body composition was measured by dual-energy X-ray absorptiometry at 14, 45, and 140 d. Serum biochemistry and glucose-insulin values (after a fasting intravenous glucose tolerance test) were obtained at 140 d. Liver and muscle were analyzed for insulin receptor signaling and triglycerides. RESULTS Body weight was similar, but percent fat was higher, whereas percent lean and bone mineral density were lower in TPN than in EN pigs (P < 0.01) at 45 d of age but not at 140 d. At 140 d, there were no differences in serum markers of liver injury or lipidemia. Intravenous glucose tolerance test at 140 d showed a lower (P < 0.05) AUC for both glucose and insulin in TPN than in EN pigs, but the ratio of AUCs of insulin and glucose was not different between groups. CONCLUSIONS Administration of TPN during the neonatal period increased adipose deposition that transiently persisted in early adolescence when challenged with a high-fat diet but was not sustained or manifested as glucose intolerance.
Collapse
Affiliation(s)
- Sarah K Elefson
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Barbara Stoll
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Teresa A Davis
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Marta L Fiorotto
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Samer W El-Kadi
- School of Animal Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Kenneth Genovese
- USDA Southern Plains Agricultural Research Center, College Station, TX, United States
| | - Thomas Thymann
- Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Per T Sangild
- Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Douglas G Burrin
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
4
|
Serrano-Jara D, Rivera-Gomis J, Tornel JA, Jordán MJ, Martínez-Conesa C, Pablo MJC. Oregano Essential Oil and Purple Garlic Powder Effects on Intestinal Health, Microbiota Indicators and Antimicrobial Resistance as Feed Additives in Weaning Piglets. Animals (Basel) 2023; 14:111. [PMID: 38200842 PMCID: PMC10778277 DOI: 10.3390/ani14010111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Finding alternatives to zinc oxide is a pressing issue for the pig production sector. We studied the impact of the bioactive components degradation of oregano essential oil (OEO) and purple garlic powder (PGP) during storage in silos, their effect on the morphometry of the jejunum and ileum and the cecal microbiota as intestinal health indicators in piglets during the post-weaning period. We also monitored antimicrobial resistance in the commensal indicator E. coli. Histological parameters and intestinal microbiota were measured in 140 piglets weaned at 21 days of age. Seven dietary treatments were used: a negative control group (basal diet), a positive control group with ZnO (3000 mg/kg of food), two groups with OEO at 0.4% and 1.2%, respectively, two groups with PGP 0.4% and 2%, respectively, and a group with OEO with 1.2% combined with PGP with 2%. Each group of piglets received the treatment for seven weeks, from weaning, before samples were taken. Antibiotic resistance profiles were measured in 81 E. coli strains. On this occasion, only the control groups, ZnO, OEO 1.2%, PGP 2% and OEO 1.2% + PGP 2% were used, and the samples were obtained from the cecal content. A progressive loss of the bioactive components of OEO and PGP was observed during the 34 days of storage (p < 0.05). PGP 2%, OEO 1.2% and their combination showed results similar to ZnO (p > 0.05), or superior in the study of intestinal morphometry and the values of E. coli and Lactobacillus. All categories showed high levels of resistance. Only the strains isolated from the OEO 1.2% group did not show resistance to colistin and presented the lowest resistance values. In general, high doses of the additives studied showed the best results, obtaining levels like or higher than those offered by ZnO.
Collapse
Affiliation(s)
- Daniel Serrano-Jara
- Department of Comparative Anatomy and Pathology, Veterinary Medicine Faculty, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, Espinardo, 30100 Murcia, Spain;
| | - Jorge Rivera-Gomis
- Scotland’s Rural College (SRUC), Centre for Epidemiology and Planetary Health, Inverness, Scotland IV2 5NA, UK
| | | | - María José Jordán
- Research Group on Rainfed Agriculture for Rural Development, Department of Rural Development, Oenology and Sustainable Agriculture, Murcia Institute of Agri-Food and Environmental Research (IMIDA), La Alberca de Las Torres, 30150 Murcia, Spain; (M.J.J.); (C.M.-C.)
| | - Cristina Martínez-Conesa
- Research Group on Rainfed Agriculture for Rural Development, Department of Rural Development, Oenology and Sustainable Agriculture, Murcia Institute of Agri-Food and Environmental Research (IMIDA), La Alberca de Las Torres, 30150 Murcia, Spain; (M.J.J.); (C.M.-C.)
| | - María José Cubero Pablo
- Animal Health Department, Veterinary Medicine Faculty, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, Espinardo, 30100 Murcia, Spain;
| |
Collapse
|
5
|
Prahm AP, Hvistendahl MK, Brandt CF, Blanche P, Hartmann B, Holst JJ, Jeppesen PB. Post-prandial secretion of glucagon-like peptide-2 (GLP-2) after carbohydrate-, fat- or protein enriched meals in healthy subjects. Peptides 2023; 169:171091. [PMID: 37640265 DOI: 10.1016/j.peptides.2023.171091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/15/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Glucagon-like peptide 2 (GLP-2) is an important regulator of intestinal growth and function. In adherable mixed meals the macronutrient composition with the best potential for stimulating GLP-2 secretion is not known. We compared the effect of 3 iso-energetic meals, where approximately 60 % of the energy ratio was provided as either carbohydrate, fat, or protein, respectively, on the post-prandial endogenous GLP-2 secretion. The responses were compared to secretion profiles of peptide YY (PYY), and glucose-dependent insulinotropic peptide (GIP). Ten healthy subjects were admitted on three occasions, at least a week apart, after a night of fasting. In an open-label, crossover design, they were randomized to receive a high carbohydrate (HC), high fat (HF) or high protein (HP) meal. The meals were approximately ∼3.9 MJ. Venous blood was collected for 240 min, and plasma concentrations of GLP-2, GIP and PYY were measured with specific radioimmunoassays. Mean GLP-2 levels peaked already at 30 min for the HC meal, however the HP meal induced the highest mean GLP-2 peaking levels, resulting in significantly higher mean GLP-2 area under the curve (AUC) from baseline of 7279 pmol*min/L, 95 %-CI [6081;8477] compared to the HC meal: 4764 pmol*min/L, 95 %-CI [3498;6029], p = 0.020 and the HF meal: 4796 pmol*min/L, [3385;6207], p = 0.011. Findings were similar for the PYY. The HC meal provided a greater AUC for GIP compared to the HP- and HF meals. The HP meal was most effective with respect to stimulation of the postprandial GLP-2 and PYY secretion, whereas the HC meal was more effective for GIP.
Collapse
Affiliation(s)
- August Pilegaard Prahm
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Mark Krogh Hvistendahl
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Christopher Filtenborg Brandt
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Paul Blanche
- Section of Biostatistics, University of Copenhagen, Øster Farimagsgade 5, 1353 Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences and the NovoNordisk Foundation, Center for Basic Metabolic Research, Faculty of Health Science, Panum Institute 12.2, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences and the NovoNordisk Foundation, Center for Basic Metabolic Research, Faculty of Health Science, Panum Institute 12.2, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Palle Bekker Jeppesen
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
6
|
Liermann W, Tümmler LM, Kuhla B, Viergutz T, Hammon HM. Effects of rumen cannulation combined with different pre-weaning feeding intensities on the intestinal, splenic and thymic immune system in heifer calves several month after surgery. Front Immunol 2023; 14:1160935. [PMID: 37143684 PMCID: PMC10151785 DOI: 10.3389/fimmu.2023.1160935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Fistulation is a helpful procedure in animal nutritional research and also common practise in human medicine. However, there are indications that alterations in the upper gastrointestinal tract contribute to intestinal immune modulations. The present study aimed to investigate effects of a rumen cannulation in week 3 of life on the intestinal and tissue specific immune system of 34-week old heifers. Nutrition influences the development of the neonatal intestinal immune system to a high extent. Therefore, rumen cannulation was investigated in combination with different pre-weaning milk feeding intensities (20% (20MR) vs. 10% milk replacer feeding (10MR). Heifers of 20MR without rumen cannula (NRC) showed higher cluster of differentiation (CD)8+ T cell subsets in mesenteric lymph nodes (MSL) compared to heifers with rumen cannula (RC) and 10MRNRC heifers. CD4+ T cell subsets in jejunal intraepithelial lymphocytes (IELs) were higher in 10MRNRC heifers compared to 10MRRC heifers. CD4+ T cell subsets in ileal IELs were lower and CD21+ B cell subsets were higher in NRC heifers compared to RC heifers. CD8+ T cell subsets in spleen tended to be lower in 20MRNRC heifers compared to all other groups. Splenic CD21+ B cell subsets were higher in 20MRNRC heifers compared to RC heifers. Splenic toll like receptor (TLR) 6 expression was increased and IL4 expression tended to be increased in RC heifers than NRC heifers. Splenic TLR2, 3 and 10 gene expression was higher in 20MR compared to 10MR heifers. Jejunal prostaglandin endoperoxide synthase 2 expression was higher in RC heifers than NRC heifers, and MUC2 expression tended to increase in 20MR heifers compared to 10MR heifers. In conclusion, rumen cannulation modulated T and B cell subsets in the down streaming gastrointestinal tract and spleen. Pre-weaning feeding intensity seemed to affect intestinal mucin secretion and T and B cell subsets in MSL, spleen and thymus until several month later. Interestingly, in MSL, spleen and thymus the 10MR feeding regime evoked similar modulations of T and B cell subsets like rumen cannulation.
Collapse
Affiliation(s)
- Wendy Liermann
- Institute of Nutritional Physiology "Oskar Kellner", Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- *Correspondence: Wendy Liermann,
| | - Lisa-Maria Tümmler
- Institute of Nutritional Physiology "Oskar Kellner", Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Björn Kuhla
- Institute of Nutritional Physiology "Oskar Kellner", Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Torsten Viergutz
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Harald Michael Hammon
- Institute of Nutritional Physiology "Oskar Kellner", Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
7
|
Kirupananthan D, Bertolo RF, Brunton JA. Lysine Dipeptide Enhances Gut Structure and Whole-Body Protein Synthesis in Neonatal Piglets with Intestinal Atrophy. J Nutr 2022; 152:1843-1850. [PMID: 35481706 DOI: 10.1093/jn/nxac095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/28/2022] [Accepted: 04/21/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Parenteral nutrition (PN) is often a necessity for preterm infants; however, prolonged PN leads to gut atrophy, weakened gut barrier function, and a higher risk of intestinal infections. Peptide transporter-1 (PepT1) is a di- or tripeptide transporter in the gut and, unlike other nutrient transporters, its activity is preserved with the onset of intestinal atrophy from PN. As such, enteral amino acids in the form of dipeptides may be more bioavailable than free amino acids when atrophy is present. OBJECTIVES In Yucatan miniature piglets with PN-induced intestinal atrophy, we sought to determine the structural and functional effects of enteral refeeding with lysine as a dipeptide, compared to free L-lysine. METHODS Piglets aged 7-8 days were PN-fed for 4 days to induce intestinal atrophy, then were refed with enteral diets with equimolar lysine supplied as lysyl-lysine (Lys-Lys; n = 7), free lysine (n = 7), or Lys-Lys with glycyl-sarcosine (n = 6; to determine whether competitive inhibition of Lys-Lys uptake would abolish PepT1-mediated effects). The diets provided lysine at 75% of the requirement and were gastrically delivered for a total of 18 hours. Whole-body and tissue-specific protein synthesis, as well as indices for gut structure and barrier function, were measured. RESULTS The villus height, mucosal weight, and free lysine concentration were higher in the Lys-Lys group compared to the other 2 groups (P < 0.05). Lysyl-lysine led to greater whole-body protein synthesis compared to free lysine (P < 0.05). Mucosal myeloperoxidase activity was lower in the Lys-Lys group (P < 0.05), suggesting less inflammation. The inclusion of glycyl-sarcosine with Lys-Lys abolished the dipeptide effects on whole-body and tissue-specific protein synthesis (P < 0.05), suggesting that improved lysine availability was mediated by PepT1. CONCLUSIONS Improved intestinal structure and whole-body protein synthesis suggests that feeding strategies designed to exploit PepT1 may help to avoid adverse effects when enteral nutrition is reintroduced into the compromised guts of neonatal piglets.
Collapse
Affiliation(s)
| | - Robert F Bertolo
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Janet A Brunton
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
8
|
Hinchliffe T, Pauline ML, Wizzard PR, Jovel J, Nation PN, Wales PW, Madsen KL, Turner JM. The effect of fecal microbial transplant on intestinal microbial composition in short bowel neonatal piglets. JPEN J Parenter Enteral Nutr 2022; 46:1393-1403. [PMID: 35043436 DOI: 10.1002/jpen.2333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/29/2021] [Accepted: 01/03/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Short bowel syndrome (SBS) in neonates is associated with microbial dysbiosis due to intestinal surgery, prolonged hospitalization, enteral nutrition, and repeated antibiotic exposure. Sepsis and liver disease, leading causes of morbidity and mortality in SBS, may relate to such intestinal dysbiosis. We investigated the safety and feasibility of fecal microbial transplant (FMT) to alter intestinal microbial composition in SBS piglets. METHODS Following a 75% distal small intestinal resection, piglets were fed parenteral nutrition (PN) and elemental diet (ED), and randomized to saline (SAL, n=12) or FMT (n=12) treatments delivered by gastric tube on day 2 (d2). FMT donor was a healthy adult pig. Comparisons were also made to healthy sow-fed littermate controls (SOW, n=6). Stool samples were collected daily, and tissue samples were collected at baseline and termination. Microbial DNA was extracted from stool and analyzed using 16S rRNA sequencing. RESULTS All piglets survived to the endpoint. On d2-4, FMT piglets had some differences in microbiota composition, compared to SAL, SOW, and donor. Between base and term, there were transitory changes to alpha and beta diversity in FMT and SAL. CONCLUSION FMT treatment in post-surgical neonatal piglets with SBS appears safe, with no increase in sepsis and no mortality. In SBS piglets, FMT induced transient changes to the intestinal microbiota. However, these changes did not persist long-term. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tierah Hinchliffe
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Mirielle L Pauline
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela R Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Juan Jovel
- Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick N Nation
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Paul W Wales
- Department of Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati
| | - Karen L Madsen
- Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Justine M Turner
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Patel JJ, Shukla A, Heyland DK. Enteral nutrition in septic shock: A pathophysiologic conundrum. JPEN J Parenter Enteral Nutr 2021; 45:74-78. [PMID: 34897735 DOI: 10.1002/jpen.2246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/02/2021] [Accepted: 08/02/2021] [Indexed: 11/07/2022]
Abstract
Septic shock is a public health burden and defined as a subset of sepsis whereby abnormalities in microcirculatory and cellular metabolism manifest as acute circulatory failure. At the level of the gut, septic shock impairs epithelial barrier function (EBF), and the gut initiates proinflammatory responses contributing to multiple organ dysfunction syndrome. The timing and dose of enteral nutrition (EN) in septic shock remains a conundrum. On the one hand, early EN preserves EBF. On the other hand, serious gastrointestinal complications such as bowel necrosis may limit EN initiation in septic shock. We (1) describe the pathophysiologic conundrum septic shock poses for EN initiation, (2) outline guideline-based recommendations for EN in septic shock, (3) identify the role of parenteral nutrition in septic shock, and (4) identify and appraise postguideline literature on the timing, dose, and titration of EN in septic shock.
Collapse
Affiliation(s)
- Jayshil J Patel
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Anuj Shukla
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Daren K Heyland
- Department of Critical Care Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
10
|
Cantet JM, Yu Z, Ríus AG. Heat Stress-Mediated Activation of Immune-Inflammatory Pathways. Antibiotics (Basel) 2021; 10:antibiotics10111285. [PMID: 34827223 PMCID: PMC8615052 DOI: 10.3390/antibiotics10111285] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 12/23/2022] Open
Abstract
Physiological changes in animals exposed to elevated ambient temperature are characterized by the redistribution of blood toward the periphery to dissipate heat, with a consequent decline in blood flow and oxygen and nutrient supply to splanchnic tissues. Metabolic adaptations and gut dysfunction lead to oxidative stress, translocation of lumen contents, and release of proinflammatory mediators, activating a systemic inflammatory response. This review discusses the activation and development of the inflammatory response in heat-stressed models.
Collapse
|
11
|
The Farnesoid X Receptor Agonist Tropifexor Prevents Liver Damage in Parenteral Nutrition-fed Neonatal Piglets. J Pediatr Gastroenterol Nutr 2021; 73:e11-e19. [PMID: 33783400 DOI: 10.1097/mpg.0000000000003135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Intestinal failure-associated liver disease (IFALD) is a life-threatening complication for patients with intestinal failure who receive long-term parenteral nutrition (PN). We evaluated the effects of the farnesoid X receptor agonist tropifexor on a neonatal piglet model of IFALD fed with PN. METHODS The piglets received PN and tropifexor for 14 days, then levels of liver enzymes, bile acid metabolism, inflammation, and intestinal barrier markers were assessed using quantitative real-time PCR. Fibroblast growth factor (FGF) 19 serum levels were determined using enzyme-linked immunosorbent assays. Bile acids were determined in liver, serum, and intestinal contents, and the microbiome was sequenced in different intestinal segments. RESULTS The PN model was established in newborn piglets. The levels of serum liver enzymes, pro-inflammatory factors, and oxidative stress increased in the livers of piglets fed with PN, but not in those fed with PN and tropifexor. Tropifexor stimulated FGF19 expression in ileal epithelial cells, increased portal FGF19 levels, then inhibited cholesterol 7α-hydroxylase expression in the liver. Tropifexor increased the relative abundance of bacteria associated with bile salt hydrolase and 7α-dehydrogenation in the contents of ileum and altered the composition of bile acids in serum, liver, and intestinal contents. Tropifexor also inhibited intestinal inflammation, alleviated intestinal mucosal atrophy, and improved the intestinal barrier. CONCLUSIONS Tropifexor might prevent liver damage in neonatal piglets receiving PN by altering the composition of intestinal microbiota and bile acids. Tropifexor also alleviates intestinal inflammation and preserves the intestinal barrier.
Collapse
|
12
|
Kelly P. Starvation and Its Effects on the Gut. Adv Nutr 2021; 12:897-903. [PMID: 33271592 PMCID: PMC8166558 DOI: 10.1093/advances/nmaa135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/09/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
There is growing awareness that intestinal dysfunction determines the clinical outcomes of situations as diverse as undernourished children in urban tropical slums and undernourished surgical patients in intensive care units. As experimental starvation in humans has only rarely been studied, and largely not using current biomedical research tools, we must draw inference from disparate clinical and experimental observations as to the derangements present in the starved gut. There is good evidence of intestinal atrophy and achlorhydria in starvation and severe undernutrition. Historical reports from concentration camps and conflict settings consistently reported a noncontagious phenomenon called "hunger diarrhea," but in settings where starved individuals are isolated from others (prisoners on hunger strike, anorexia nervosa) diarrhea is not a feature. Changes in intestinal permeability and absorption have been infrequently studied in experimental starvation; available data suggest that short-term starvation reduces sugar absorption but not permeability. Severe acute malnutrition in children is associated with severe changes in the intestinal mucosa. Experimental animal models may help explain some observations in humans. Starved rats develop a hypersecretory state and intestinal barrier defects. Starved pigs demonstrate prolongation of rotavirus diarrhea and reproduce some of the absorptive and barrier defects observed in malnourished children. However, there remains much to be learned about the effects of starvation on the gut. Given the high prevalence of undernutrition in hospitals and disadvantaged communities, the lack of attention to the interaction between undernutrition and gastrointestinal damage is surprising and needs to be corrected. Current sophisticated cellular and molecular techniques now provide the opportunity to create fresh understanding of gastrointestinal changes in pure undernutrition, using volunteer studies and samples from anorexia nervosa.
Collapse
Affiliation(s)
- Paul Kelly
- Blizard Institute, Barts and The London School of Medicine, London, United Kingdom; and Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
| |
Collapse
|
13
|
Lansing M, Slim G, Wizzard P, Rafii M, Pencharz PB, Nation PN, Beggs MR, Alexander RT, Wales PW, Turner JM, Ball RO. Intestinal resection affects whole-body arginine synthesis in neonatal piglets. Pediatr Res 2021; 89:1420-1426. [PMID: 32920606 DOI: 10.1038/s41390-020-01139-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Previous studies in piglets show a direct relationship between intestinal mass and arginine (Arg) synthesis. We aimed to study the effects of 75% intestinal resection on whole-body Arg synthesis. METHODS Piglets were allocated to sham or jejunocolic (JC) surgery and to enteral nutrition (EN) at 20% [sham (n = 8), JC (n = 10)], or 40% [sham (n = 4), JC (n = 5)]. A gastric tube was placed for EN and a venous catheter for parenteral nutrition and blood sampling. On day 6, a primed bolus and constant infusion of Arg m + 2 label and proline m + 1 label was delivered. In addition, 40% EN piglets received a citrulline (Cit) m + 3 tracer. Blood sampling was undertaken and whole-body Arg synthesis was calculated. On day 7, intestinal length was measured, and samples were collected for gene expression (PCR quantification) and histopathology. RESULTS On Day 7, sham piglets showed intestinal lengthening compared to JC (p = 0.02). Whole-body Arg synthesis was similar between groups (p = 0.50). Adjusting for absolute small intestinal length, JC piglets had greater Arg synthesis (p = 0.01). Expression of arginosuccinase was upregulated in the jejunum of JC compared to sham on 20% EN (p = 0.03). CONCLUSION This demonstrates for the first-time adaptive changes in intestinal Arg synthesis following intestinal resection. IMPACT The intestine makes a critical contribution to whole-body arginine synthesis, particularly in neonates, a human population at risk for short bowel syndrome. Therefore, we studied intestinal arginine synthesis in a neonatal piglet model of short bowel syndrome and demonstrated adaptive changes in the intestine that may preserve whole-body arginine synthesis, despite loss of intestinal mass. This research adds new information to our understanding of the effects a massive intestinal resection has on amino acid metabolism during neonatal development.
Collapse
Affiliation(s)
- Marihan Lansing
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - George Slim
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Pamela Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Mahroukh Rafii
- Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Paul B Pencharz
- Departments of Pediatrics and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Patrick N Nation
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Megan R Beggs
- Department of Physiology at the University of Alberta, Edmonton, AB, Canada
| | - R Todd Alexander
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Physiology at the University of Alberta, Edmonton, AB, Canada
| | - Paul W Wales
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Justine M Turner
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
| | - Ron O Ball
- Department of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Ayuso M, Irwin R, Walsh C, Van Cruchten S, Van Ginneken C. Low birth weight female piglets show altered intestinal development, gene expression, and epigenetic changes at key developmental loci. FASEB J 2021; 35:e21522. [PMID: 33734504 DOI: 10.1096/fj.202002587r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/15/2021] [Accepted: 02/26/2021] [Indexed: 12/15/2022]
Abstract
Intestinal development is compromised in low birth weight (LBW) pigs, negatively impacting their growth, health, and resilience. We investigated the molecular mechanisms of the altered intestinal maturation observed in neonatal and juvenile LBW female piglets by comparing the changes in intestinal morphology, gene expression, and methylation in LBW versus normal birth weight (NBW) female piglets. A total of 16 LBW/NBW sibling pairs were sacrificed at 0 hours, 8 hours, 10 days, and 8 weeks of age. The gastrointestinal tract was weighed, measured, and the small intestine was sampled for histomorphology, gene expression, and methylation analyses. Impaired intestinal development, with shorter villi and shallower crypts, was observed in LBW female piglets. The expression of intestinal development markers (ALPI and OLFM) rapidly peaked after birth in NBW but not in LBW female piglets. The lower expression of genes involved in nutrient digestion (ANPEP and SI) and barrier function (OCLN and CLDN4) in LBW, together with their delayed development of intestinal villi and crypts could help to explain the compromised health and growth potential of LBW female piglets. The changes in methylation observed in LBW in key regulators of intestinal development (OLFM4 and FZD5) suggest long-term effects of BW on intestinal gene expression, development, and function. Accordingly, experimental demethylation induced in IPEC-J2 cells led to increased expression of intestinal genes (MGA, DPP4, and GLUT2). Overall, we have identified the alterations in transcription or epigenetic marking at a number of genes critical to intestinal development, which may contribute to both the short- and long-term failure of LBW female piglets to thrive.
Collapse
Affiliation(s)
- Miriam Ayuso
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Rachelle Irwin
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - Colum Walsh
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - Steven Van Cruchten
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Chris Van Ginneken
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
15
|
El-Kadi SW, Boutry-Regard C, Suryawan A, Nguyen HV, Kimball SR, Fiorotto ML, Davis TA. Intermittent Bolus Feeding Enhances Organ Growth More Than Continuous Feeding in a Neonatal Piglet Model. Curr Dev Nutr 2020; 4:nzaa170. [PMID: 33381676 PMCID: PMC7751947 DOI: 10.1093/cdn/nzaa170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Orogastric tube feeding is frequently prescribed for neonates who cannot ingest food normally. In a piglet model of the neonate, greater skeletal muscle growth is sustained by upregulation of translation initiation signaling when nutrition is delivered by intermittent bolus meals, rather than continuously. OBJECTIVES The objective of this study was to determine the long-term effects of feeding frequency on organ growth and the mechanism by which feeding frequency modulates protein anabolism in these organs. METHODS Eighteen neonatal pigs were fed by gastrostomy tube the same amount of a sow milk replacer either by continuous infusion (CON) or on an intermittent bolus schedule (INT). After 21 d of feeding, the pigs were killed without interruption of feeding (CON; n = 6) or immediately before (INT-0; n = 6) or 60 min after (INT-60; n = 6) a meal, and fractional protein synthesis rates and activation indexes of signaling pathways that regulate translation initiation were measured in the heart, jejunum, ileum, kidneys, and liver. RESULTS Compared with continuous feeding, intermittent feeding stimulated the growth of the liver (+64%), jejunum (+48%), ileum (+40%), heart (+64%), and kidney (+56%). The increases in heart, kidney, jejunum, and ileum masses were proportional to whole body lean weight gain, but liver weight gain was greater in the INT-60 than the CON, and intermediate for the INT-0 group. For the liver and ileum, but not the heart, kidney, and jejunum, INT-60 compared with CON pigs had greater fractional protein synthesis rates (22% and 48%, respectively) and was accompanied by an increase in ribosomal protein S6 kinase 1 and eukaryotic initiation factor 4E binding protein 1 phosphorylation. CONCLUSIONS These results suggest that intermittent bolus compared with continuous orogastric feeding enhances organ growth and that in the ileum and liver, intermittent feeding enhances protein synthesis by stimulating translation initiation.
Collapse
Affiliation(s)
- Samer W El-Kadi
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Claire Boutry-Regard
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Agus Suryawan
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hanh V Nguyen
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Marta L Fiorotto
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Teresa A Davis
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
16
|
Hinchliffe T, Pauline ML, Wizzard PR, Nation PN, Brubaker P, Campbell JR, Kim Y, Dimitriadou V, Wales PW, Turner JM. Durability of Linear Small-Intestinal Growth Following Treatment Discontinuation of Long-Acting Glucagon-Like Peptide 2 (GLP-2) Analogues. JPEN J Parenter Enteral Nutr 2020; 45:1466-1474. [PMID: 33241564 DOI: 10.1002/jpen.2053] [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: 07/26/2020] [Revised: 10/23/2020] [Accepted: 11/17/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Short-bowel syndrome is the leading cause of pediatric intestinal failure, resulting in dependency on long-term parenteral nutrition (PN). To promote enteral autonomy in neonates, a key outcome may be intestinal growth in length. The purpose of this study was to determine if intestinal lengthening persists following discontinuation of treatment with 1 of 2 GLP-2 analogues with different pharmacokinetic profiles. METHODS Neonatal short-bowel piglets were assigned to saline control (S), 7-day treatment with teduglutide (T) (0.05 mg/kg twice daily), or 7-day treatment with apraglutide (A) (5 mg/kg twice weekly). Comparisons were made between day 7 and day 14 endpoints using analysis of variance. Data included small-intestine length, weight, histology, and quantitative polymerase chain reaction analysis of mucosal transcripts for peptide growth factors and their receptors, nutrient transporters, and tight-junction proteins. RESULTS Compared with control, 7 days of GLP-2 analogue treatment induced mucosal adaptation based on villus hyperplasia (P = .003), which was not durable 7 days after treatment cessation (day 14; P = .081). Treatment increased intestinal growth in length by day 7 (P = .005), which was maintained (by T) or further increased (by A) at day 14 (P < .001). No significant differences in mucosal transcripts were detected. CONCLUSION Unlike mucosal adaptation, intestinal growth appears to be a lasting outcome of treatment with long-acting GLP-2 analogues in a neonatal piglet short-bowel model. This has significant clinical implications for neonates, given their potential for intestinal growth. Intestinal lengthening varies between analogues with different half-lives; however, molecular mechanisms require further elucidation.
Collapse
Affiliation(s)
- Tierah Hinchliffe
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Mirielle L Pauline
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela R Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick N Nation
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Patricia Brubaker
- Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jhenielle R Campbell
- Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yunji Kim
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | | - Paul W Wales
- Department of Surgery, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Justine M Turner
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Koepsell H. Glucose transporters in the small intestine in health and disease. Pflugers Arch 2020; 472:1207-1248. [PMID: 32829466 PMCID: PMC7462918 DOI: 10.1007/s00424-020-02439-5] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 12/23/2022]
Abstract
Absorption of monosaccharides is mainly mediated by Na+-D-glucose cotransporter SGLT1 and the facititative transporters GLUT2 and GLUT5. SGLT1 and GLUT2 are relevant for absorption of D-glucose and D-galactose while GLUT5 is relevant for D-fructose absorption. SGLT1 and GLUT5 are constantly localized in the brush border membrane (BBM) of enterocytes, whereas GLUT2 is localized in the basolateral membrane (BLM) or the BBM plus BLM at low and high luminal D-glucose concentrations, respectively. At high luminal D-glucose, the abundance SGLT1 in the BBM is increased. Hence, D-glucose absorption at low luminal glucose is mediated via SGLT1 in the BBM and GLUT2 in the BLM whereas high-capacity D-glucose absorption at high luminal glucose is mediated by SGLT1 plus GLUT2 in the BBM and GLUT2 in the BLM. The review describes functions and regulations of SGLT1, GLUT2, and GLUT5 in the small intestine including diurnal variations and carbohydrate-dependent regulations. Also, the roles of SGLT1 and GLUT2 for secretion of enterohormones are discussed. Furthermore, diseases are described that are caused by malfunctions of small intestinal monosaccharide transporters, such as glucose-galactose malabsorption, Fanconi syndrome, and fructose intolerance. Moreover, it is reported how diabetes, small intestinal inflammation, parental nutrition, bariatric surgery, and metformin treatment affect expression of monosaccharide transporters in the small intestine. Finally, food components that decrease D-glucose absorption and drugs in development that inhibit or downregulate SGLT1 in the small intestine are compiled. Models for regulations and combined functions of glucose transporters, and for interplay between D-fructose transport and metabolism, are discussed.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstr 6, 97070, Würzburg, Germany.
| |
Collapse
|
18
|
Hargrove DM, Alagarsamy S, Croston G, Laporte R, Qi S, Srinivasan K, Sueiras-Diaz J, Wiśniewski K, Hartwig J, Lu M, Posch AP, Wiśniewska H, Schteingart CD, Rivière PJM, Dimitriadou V. Pharmacological Characterization of Apraglutide, a Novel Long-Acting Peptidic Glucagon-Like Peptide-2 Agonist, for the Treatment of Short Bowel Syndrome. J Pharmacol Exp Ther 2020; 373:193-203. [PMID: 32075870 DOI: 10.1124/jpet.119.262238] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/18/2020] [Indexed: 12/15/2022] Open
Abstract
Glucagon-like peptide-2 (GLP-2) agonists have therapeutic potential in clinical indications in which the integrity or absorptive function of the intestinal mucosa is compromised, such as in short bowel syndrome (SBS). Native hGLP-2, a 33-amino acid peptide secreted from the small intestine, contributes to nutritional absorption but has a very short half-life because of enzymatic cleavage and renal clearance and thus is of limited therapeutic value. The GLP-2 analog teduglutide (Revestive/Gattex; Shire Inc.) has been approved for use in SBS since 2012 but has a once-daily injection regimen. Pharmacokinetic (PK) and pharmacodynamic studies confirm that apraglutide, a novel GLP-2 analog, has very low clearance, long elimination half-life, and high plasma protein binding compared with GLP-2 analogs teduglutide and glepaglutide. Apraglutide and teduglutide retain potency and selectivity at the GLP-2 receptor comparable to native hGLP-2, whereas glepaglutide was less potent and less selective. In rat intravenous PK studies, hGLP-2, teduglutide, glepaglutide, and apraglutide had clearances of 25, 9.9, 2.8, and 0.27 ml/kg per minute, respectively, and elimination half-lives of 6.4, 19, 16, and 159 minutes, respectively. The unique PK profile of apraglutide administered via intravenous and subcutaneous routes was confirmed in monkey and minipig and translated into significantly greater in vivo pharmacodynamic activity, measured as small intestinal growth in rats. Apraglutide showed greater intestinotrophic activity than the other peptides when administered at less-frequent dosing intervals because of its prolonged half-life. We postulate that apraglutide offers several advantages over existing GLP-2 analogs and is an excellent candidate for the treatment of gastrointestinal diseases, such as SBS. SIGNIFICANCE STATEMENT: Apraglutide is a potent and selective GLP-2 agonist with an extremely low clearance and prolonged elimination half-life, which differentiates it from teduglutide (the only approved GLP-2 agonist). The enhanced pharmacokinetics of apraglutide will benefit patients by enabling a reduced dosing frequency and removing the need for daily injections.
Collapse
Affiliation(s)
- Diane M Hargrove
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Sudarkodi Alagarsamy
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Glenn Croston
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Régent Laporte
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Steve Qi
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Karthik Srinivasan
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Javier Sueiras-Diaz
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Kazimierz Wiśniewski
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Jennifer Hartwig
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Mark Lu
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Alexander P Posch
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Halina Wiśniewska
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Claudio D Schteingart
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Pierre J-M Rivière
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| | - Violetta Dimitriadou
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California (D.M.H., S.A., G.C., R.L., S.Q., K.S., J.S.-D., K.W., J.H., M.L., A.P.P., H.W., C.D.S., P.J.-M.R.) and VectivBio AG, Basel, Switzerland (V.D.)
| |
Collapse
|
19
|
Pyo J, Hare K, Pletts S, Inabu Y, Haines D, Sugino T, Guan LL, Steele M. Feeding colostrum or a 1:1 colostrum:milk mixture for 3 days postnatal increases small intestinal development and minimally influences plasma glucagon-like peptide-2 and serum insulin-like growth factor-1 concentrations in Holstein bull calves. J Dairy Sci 2020; 103:4236-4251. [PMID: 32171512 DOI: 10.3168/jds.2019-17219] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 01/05/2020] [Indexed: 11/19/2022]
Abstract
This study evaluated how feeding colostrum- or a colostrum-milk mixture for 3 d postnatal affects plasma glucagon-like peptide-2 (GLP-2), serum insulin-like growth factor-1 (IGF-1), and small intestinal histomorphology in calves. Holstein bulls (n = 24) were fed colostrum at 2 h postnatal and randomly assigned to receive either colostrum (COL), whole milk (WM), or a 1:1 COL:WM mixture (MIX) every 12 h from 12 to 72 h. A jugular venous catheter was placed at 1 h postnatal to sample blood frequently for the duration of the experiment. Samples were collected at 1, 2, 3, 6, 9, 11, and 12 h. Following the 12-h meal, blood was collected at half-hour intervals until 16 h and then at 1-h intervals from 16 to 24 h. A 27-h sample was taken, then blood was sampled every 6 h from 30 to 60 h. Again, blood was taken at half-intervals from 60 to 64 h, then at 65 and 66 h, following which, a 2-h sampling interval was used until 72 h. Plasma GLP-2 (all time points) and serum IGF-1 (at time points: 1, 6, 12, 18, 24, 36, 48, and 72 h) were both analyzed. Duodenal, jejunal, and ileal tissues were collected at 75 h of age to assess histomorphology and cellular proliferation. Feeding COL, rather than WM, increased plasma GLP-2 by 60% for 2 h and tended to increase GLP-2 by 49.4% for 4 h after the 60-h meal. Insulin-like growth factor-1 area under the curve (from 12 to 72 h) tended to be 27% greater for COL than WM calves but was otherwise unaffected by treatment. Ileal crypts tended to proliferate more with MIX than WM, whereas ileal crypt proliferation did not differ for COL compared with MIX or WM and was not different between treatments in the proximal jejunum. Villi height was increased 1.8 and 1.5× (COL and MIX vs. WM) in the proximal and distal jejunum, respectively, whereas MIX duodenal and ileal villi height tended to be 1.5 and 1.4× that of WM. Crypt depth did not differ in any region. Surface area of the gastrointestinal tract was reduced for WM by 60 and 58% (proximal jejunum) and 38 and 52% (ileum) relative to COL and MIX and was 54% less than MIX in the distal jejunum. Overall, extended COL feeding minimally increased plasma GLP-2 and serum IGF-1 compared with WM feeding. As COL and MIX similarly promoted small intestinal maturation, feeding calves transition milk to promote intestinal development could be a strategy for producers.
Collapse
Affiliation(s)
- J Pyo
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, Canada T6G 2P5
| | - K Hare
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, Canada N1G 1Y2
| | - S Pletts
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, Canada T6G 2P5
| | - Y Inabu
- The Research Center for Animal Science, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Japan 739-8528
| | - D Haines
- The Saskatoon Colostrum Company Ltd., Saskatoon, SK, Canada S7K 6A2
| | - T Sugino
- The Research Center for Animal Science, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Japan 739-8528
| | - L L Guan
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, Canada T6G 2P5
| | - M Steele
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, Canada N1G 1Y2.
| |
Collapse
|
20
|
Burrin D, Sangild PT, Stoll B, Thymann T, Buddington R, Marini J, Olutoye O, Shulman RJ. Translational Advances in Pediatric Nutrition and Gastroenterology: New Insights from Pig Models. Annu Rev Anim Biosci 2020; 8:321-354. [PMID: 32069436 DOI: 10.1146/annurev-animal-020518-115142] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pigs are increasingly important animals for modeling human pediatric nutrition and gastroenterology and complementing mechanistic studies in rodents. The comparative advantages in size and physiology of the neonatal pig have led to new translational and clinically relevant models of important diseases of the gastrointestinal tract and liver in premature infants. Studies in pigs have established the essential roles of prematurity, microbial colonization, and enteral nutrition in the pathogenesis of necrotizing enterocolitis. Studies in neonatal pigs have demonstrated the intestinal trophic effects of akey gut hormone, glucagon-like peptide 2 (GLP-2), and its role in the intestinal adaptation process and efficacy in the treatment of short bowel syndrome. Further, pigs have been instrumental in elucidating the physiology of parenteral nutrition-associated liver disease and the means by which phytosterols, fibroblast growth factor 19, and a new generation of lipid emulsions may modify disease. The premature pig will continue to be a valuable model in the development of optimal infant diets (donor human milk, colostrum), specific milk bioactives (arginine, growth factors), gut microbiota modifiers (pre-, pro-, and antibiotics), pharmaceutical drugs (GLP-2 analogs, FXR agonists), and novel diagnostic tools (near-infrared spectroscopy) to prevent and treat these pediatric diseases.
Collapse
Affiliation(s)
- Douglas Burrin
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Barbara Stoll
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Randal Buddington
- College of Nursing, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Juan Marini
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
- Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Robert J Shulman
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
21
|
Oikawa K, Sakurai M, Murakawa T, Kidokoro R, Nakano Y, Asai H, Ochiai H, Shirasawa T, Yoshimoto T, Minoura A, Kokaze A, Mizuno K. Survey of a nutrition management method for very low birthweight infants: Status before wide use of breast milk banks in Japan. Pediatr Int 2020; 62:180-188. [PMID: 31793734 PMCID: PMC7065243 DOI: 10.1111/ped.14074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/16/2019] [Accepted: 11/29/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND The importance of breast-feeding for very low birthweight (VLBW) infants has been pointed out. Some overseas studies suggested that the standardization of enteral nutrition (EN) leads to improved prognosis in VLBW infants. In Japan, however, physicians in charge of infants are responsible for making nutrition management decisions on an individual basis. We conducted an online survey to clarify the course of nutrition management of VLBW infants currently implemented in Japan. METHODS We mailed a notice to 300 representative neonatologists throughout Japan requesting their participation in the online survey. On the survey website, neonatologists responded to questions regarding the nutritional strategy for five birthweight groups (less than 500 g, 500-749 g, 750-999 g, 1,000-1,249 g and 1,250-1,499 g). RESULTS Responses were recieved from 137 neonatologists. The first choice for EN up to 1 week after birth was breast milk regardless of birthweight (92.0% for 1,250-1,499 g to 95.6% for 500-999 g). More than 30% of the respondents answered that they fast infants who weigh <750 g at birth or feed them with other mothers' breast milk until their own mother's milk becomes available. The lower the birthweight, the later EN is started, and the greater the number of days to establish EN. CONCLUSION The lower the birthweight, the more difficult it is to feed infants their own mother's milk and the later the EN is started. If donor milk is supplied in a stable manner, it takes fewer days to establish EN.
Collapse
Affiliation(s)
- Kosuke Oikawa
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan.,Department of Hygiene, Public Health and Preventive Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Motoichiro Sakurai
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Tetsuro Murakawa
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Reita Kidokoro
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Yuya Nakano
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Hideyuki Asai
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Hirotaka Ochiai
- Department of Hygiene, Public Health and Preventive Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takako Shirasawa
- Department of Hygiene, Public Health and Preventive Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takahiko Yoshimoto
- Department of Hygiene, Public Health and Preventive Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Akira Minoura
- Department of Hygiene, Public Health and Preventive Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Akatsuki Kokaze
- Department of Hygiene, Public Health and Preventive Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Katsumi Mizuno
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Humphrey B, Zhao J, Faris R. Review: Link between intestinal immunity and practical approaches to swine nutrition. Animal 2019; 13:2736-2744. [PMID: 31475667 DOI: 10.1017/s1751731119001861] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gaining a deeper understanding into the underlying mechanisms associated with intestinal function and immunity during the weaning transition is critical to help shed new light into applied nutrition approaches to improve piglet performance and health during this critical life-stage transition. The transient anorexia triggered at weaning leads to compromised intestinal barrier function and a localized inflammatory response. Considering barrier function, specific nutrient fractions appear to have a significant impact on the development and function of the immune and microbial systems around weaning. Understanding the specific impact of nutrients in the small intestine and hindgut is important for helping to bring more focus and consistency to nutritional approaches to support health and immunity during the weaning transition period. The challenge continues to be how to translate these modes of action into practical and scalable approaches for swine nutrition. We will focus specifically on practical nutritional approaches to influence intestinal immunity through lipid, protein and antioxidant nutrition.
Collapse
Affiliation(s)
- B Humphrey
- Provimi, 10 Nutrition Way, Brookville, OH 45309, USA
| | - J Zhao
- Cargill Animal Nutrition, 10383 165th Ave NW, Elk River, MN 55330, USA
| | - R Faris
- Cargill Animal Nutrition, 10383 165th Ave NW, Elk River, MN 55330, USA
| |
Collapse
|
23
|
Lin S, Stoll B, Robinson J, Pastor JJ, Marini JC, Ipharraguerre IR, Hartmann B, Holst JJ, Cruz S, Lau P, Olutoye O, Fang Z, Burrin DG. Differential action of TGR5 agonists on GLP-2 secretion and promotion of intestinal adaptation in a piglet short bowel model. Am J Physiol Gastrointest Liver Physiol 2019; 316:G641-G652. [PMID: 30920308 PMCID: PMC6580240 DOI: 10.1152/ajpgi.00360.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Enteroendocrine L cells and glucagon-like peptide 2 (GLP-2) secretion are activated in the intestinal adaptation process following bowel resection in patients with short bowel syndrome. We hypothesized that enteral activation of Takeda G protein-coupled receptor 5 (TGR5), expressed in enteroendocrine L cells, could augment endogenous GLP-2 secretion and the intestinal adaptation response. Our aim was to assess the efficacy of different TGR5 agonists to stimulate GLP-2 secretion and intestinal adaptation in a piglet short-bowel model. In study 1, parenterally fed neonatal pigs (n = 6/group) were gavaged with vehicle, olive extract (OE; 10 or 50 mg/kg), or ursolic acid (UA; 10 mg/kg), and plasma GLP-2 was measured for 6 h. In study 2, neonatal pigs (n = 6-8/group) were subjected to transection or 80% mid-small intestine resection and, after 2 days, assigned to treatments for 10 days as follows: 1) transection + vehicle (sham), 2) resection + vehicle (SBS), 3) resection + 30 mg UA (SBS + UA), and 4) resection + 180 mg/kg OE (SBS + OE). We measured plasma GLP-2, intestinal histology, cell proliferation, and gene expression, as well as whole body citrulline-arginine kinetics and bile acid profiles. In study 1, GLP-2 secretion was increased by UA and tended to be increased by OE. In study 2, SBS alone, but not additional treatment with either TGR5 agonist, resulted in increased mucosal thickness and crypt cell proliferation in remnant jejunum and ileum sections. SBS increased biliary and ileal concentration of bile acids and expression of inflammatory and farnesoid X receptor target genes, but these measures were suppressed by UA treatment. In conclusion, UA is an effective oral GLP-2 secretagogue in parenterally fed pigs but is not capable of augmenting GLP-2 secretion or the intestinal adaptation response after massive small bowel resection. NEW & NOTEWORTHY Therapeutic activation of endogenous glucagon-like peptide 2 (GLP-2) secretion is a promising strategy to improve intestinal adaptation in patients with short bowel syndrome. This study in neonatal pigs showed that oral supplementation with a selective Takeda G protein-coupled receptor 5 (TGR5) agonist is an effective approach to increase GLP-2 secretion. The results warrant further study to establish a more potent oral TGR5 agonist that can effectively improve intestinal adaptation in pediatric patients with SBS.
Collapse
Affiliation(s)
- Sen Lin
- Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu, Sichuan , People's Republic of China
| | - Barbara Stoll
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
| | - Jason Robinson
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
| | | | - Juan C Marini
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Ignacio R Ipharraguerre
- Lucta S.A., Montornès del Vallès, Spain
- Institute of Human Nutrition and Food Science, University of Kiel , Kiel , Germany
| | - Bolette Hartmann
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Stephanie Cruz
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Texas Children's Hospital , Houston, Texas
| | - Patricio Lau
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Texas Children's Hospital , Houston, Texas
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Texas Children's Hospital , Houston, Texas
| | - Zhengfeng Fang
- Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu, Sichuan , People's Republic of China
| | - Douglas G Burrin
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
- Section of Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
24
|
Haisan J, Oba M, Sugino T. The effects of feeding more milk on periprandial plasma glucagon-like peptide-2 concentrations in preweaning dairy calves. J Dairy Sci 2018; 101:11396-11402. [DOI: 10.3168/jds.2018-15026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/17/2018] [Indexed: 12/25/2022]
|
25
|
Nilaweera KN, Speakman JR. Regulation of intestinal growth in response to variations in energy supply and demand. Obes Rev 2018; 19 Suppl 1:61-72. [PMID: 30511508 PMCID: PMC6334514 DOI: 10.1111/obr.12780] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022]
Abstract
The growth of the intestine requires energy, which is known to be met by catabolism of ingested nutrients. Paradoxically, during whole body energy deficit including calorie restriction, the intestine grows in size. To understand how and why this happens, we reviewed data from several animal models of energetic challenge. These were bariatric surgery, cold exposure, lactation, dietary whey protein intake and calorie restriction. Notably, these challenges all reduced the adipose tissue mass, altered hypothalamic neuropeptide expression and increased intestinal size. Based on these data, we propose that the loss of energy in the adipose tissue promotes the growth of the intestine via a signalling mechanism involving the hypothalamus. We discuss possible candidates in this pathway including data showing a correlative change in intestinal (ileal) expression of the cyclin D1 gene with adipose tissue mass, adipose derived-hormone leptin and hypothalamic expression of leptin receptor and the pro-opiomelanocortin gene. The ability of the intestine to grow in size during depletion of energy stores provides a mechanism to maximize assimilation of ingested energy and in turn sustain critical functions of tissues important for survival.
Collapse
Affiliation(s)
- K N Nilaweera
- Department of Food Biosciences, Teagasc Food Research Centre, Fermoy, County Cork, Ireland
| | - J R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
26
|
Wang Q, Xiong X, Li J, Tu Q, Yang H, Yin Y. Energy metabolism in the intestinal crypt epithelial cells of piglets during the suckling period. Sci Rep 2018; 8:12948. [PMID: 30154497 PMCID: PMC6113243 DOI: 10.1038/s41598-018-31068-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 08/09/2018] [Indexed: 11/09/2022] Open
Abstract
We tested the hypothesis that energy metabolism in the intestinal crypt epithelial cells of piglets changes during the suckling period. The experiment began with 24 piglets from 8 litters (3 piglets per litter). One piglet from each litter was randomly selected and euthanized at 7, 14, or 21 d of age, respectively. Crypt cells were isolated from the mid-jejunum and protein synthesis was analyzed using isobaric tags for relative and absolute quantification. The production of proteins related to glycolysis was mainly decreased from Days 7 to 14 before increasing up to Day 21. Synthesis of proteins involved in fatty acids, amino acids (glutamate and glutamine), and citrate cycle metabolism was generally down-regulated for samples collected on Days 14 and 21 when compared with levels on Day 7. These results indicate that energy metabolism in the intestinal crypt epithelial cells changes during the suckling period. Furthermore, this pattern of metabolism varies among glucose, fatty acids, and amino acids. Therefore, these findings may be useful in efforts to regulate the intestinal development of piglets.
Collapse
Affiliation(s)
- Qiye Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410007, China
| | - Xia Xiong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Jianzhong Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410007, China
| | - Qiang Tu
- Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, School of Life Science, Shandong University, Jinan, China
| | - Huansheng Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410007, China. .,Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China.
| | - Yulong Yin
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410007, China. .,Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China.
| |
Collapse
|
27
|
Orhan A, Gögenur I, Kissow H. The Intestinotrophic Effects of Glucagon-Like Peptide-2 in Relation to Intestinal Neoplasia. J Clin Endocrinol Metab 2018; 103:2827-2837. [PMID: 29741675 DOI: 10.1210/jc.2018-00655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023]
Abstract
CONTEXT Glucagon-like peptide-2 (GLP-2) is a gastrointestinal hormone with intestinotrophic and antiapoptotic effects. The hormone's therapeutic potential in intestinal diseases and relation to intestinal neoplasia has raised great interest among researchers. This article reviews and discusses published experimental and clinical studies concerning the growth-stimulating and antiapoptotic effects of GLP-2 in relation to intestinal neoplasia. EVIDENCE ACQUISITION The data used in this narrative review were collected through literature research in PubMed using English keywords. All studies to date examining GLP-2's relation to intestinal neoplasms have been reviewed in this article, as the studies on the matter are sparse. EVIDENCE SYNTHESIS GLP-2 has been found to stimulate intestinal growth through secondary mediators and through the involvement of Akt phosphorylation. Studies on rodents have shown that exogenously administered GLP-2 increases the growth and incidence of adenomas in the colon, suggesting that GLP-2 may play an important role in the progression of intestinal tumors. Clinical studies have found that exogenous GLP-2 treatment is well tolerated for up to 30 months, but the tolerability for even longer periods of treatment has not been examined. CONCLUSION Exogenous GLP-2 is currently available as teduglutide for the treatment of short bowel syndrome. However, the association between exogenous GLP-2 treatment and intestinal neoplasia in humans has not been fully identified. This leads to a cause for concern regarding the later risk of the development or progression of intestinal tumors with long-term GLP-2 treatment. Therefore, further research regarding GLP-2's potential relation to intestinal cancers is needed.
Collapse
Affiliation(s)
- Adile Orhan
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Koege, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- NNF Center of Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
28
|
Early Serum Gut Hormone Concentrations Associated With Time to Full Enteral Feedings in Preterm Infants. J Pediatr Gastroenterol Nutr 2018; 67:97-102. [PMID: 29620597 DOI: 10.1097/mpg.0000000000001987] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES The primary objective of this study was to evaluate early postnatal serum gut hormone concentrations in preterm infants as predictors of time to full enteral feedings. The secondary objective was to identify infant characteristics and nutritional factors that modulate serum gut hormone concentrations and time to full enteral feedings. METHODS Sixty-four preterm infants less than 30 weeks of gestation were included in this retrospective cohort study. Serum gut hormone concentrations at postnatal days 0 and 7 were measured using enzyme-linked immunosorbent assays. Linear regression and mediation analyses were performed. RESULTS Median (interquartile range) serum concentrations of glucose-dependent insulinotropic peptide (GIP) and peptide YY (PYY) on postnatal day 7 were 31.3 pg/mL (18.2, 52.3) and 1181.7 pg/mL (859.0, 1650.2), respectively. GIP and PYY concentrations on day 7 were associated with days to full enteral feedings after adjustment for confounders (β = -1.1, P = 0.03; and β = -0.002, P = 0.02, respectively). Nutritional intake was correlated with serum concentrations of GIP and PYY on postnatal day 7 and time to full enteral feedings. Mediation analysis revealed that the effect of serum gut hormone concentrations on time to full enteral feedings was not fully explained by nutritional intake. Intrauterine growth restriction, mechanical ventilation on postnatal day 7, and patent ductus arteriosus treated with indomethacin were associated with longer time to full enteral feedings. CONCLUSIONS Serum concentrations of GIP and PYY on postnatal 7 are independently associated with time to full enteral feedings. The link between serum gut hormone concentrations and time to full enteral feedings is not fully mediated by nutritional factors, suggesting an independent mechanism underlying the influence of gut hormones on feeding tolerance and time to full enteral feedings.
Collapse
|
29
|
Enteral/Parenteral Nutrition in Foals and Adult Horses Practical Guidelines for the Practitioner. Vet Clin North Am Equine Pract 2018; 34:169-180. [PMID: 29426711 DOI: 10.1016/j.cveq.2017.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Nutritional support is an important adjunct to medical therapy in the sick, injured, or debilitated equine patient. What is not clear is the optimal route, composition, or amounts of support. The enteral route should be chosen whenever possible to maximize the benefits to the gastrointestinal tract and the patient as a whole. Complete or partial parenteral nutrition is most useful as a bridge during recovery and transition to enteral feeding in the horse. The reader is encouraged to consider nutritional support whether enteral or parenteral in any anorexic, chronically debilitated, or sick equine patient.
Collapse
|
30
|
Aunsholt L, Qvist N, Sangild PT, Vegge A, Stoll B, Burrin DG, Jeppesen PB, Eriksen T, Husby S, Thymann T. Minimal Enteral Nutrition to Improve Adaptation After Intestinal Resection in Piglets and Infants. JPEN J Parenter Enteral Nutr 2017; 42:446-454. [PMID: 28786308 DOI: 10.1177/0148607117690527] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 01/01/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Minimal enteral nutrition (MEN) may induce a diet-dependent stimulation of gut adaptation following intestinal resection. Bovine colostrum is rich in growth factors, and we hypothesized that MEN with colostrum would stimulate intestinal adaptation, compared with formula, and would be well tolerated in patients with short bowel syndrome. METHODS In experiment 1, 3-day-old piglets with 50% distal small intestinal resection were fed parenteral nutrition (PN, n = 10) or PN plus MEN given as either colostrum (PN-COL, n = 5) or formula (PN-FORM, n = 9) for 7 days. Intestinal nutrient absorption and histomorphometry were performed. In experiment 2, tolerance and feasibility of colostrum supplementation were tested in a pilot study on 5 infants who had undergone intestinal resection, and they were compared with 5 resected infants who served as controls. RESULTS In experiment 1, relative wet-weight absorption and intestinal villus height were higher in PN-COL vs PN (53% vs 23% and 362 ± 13 vs 329 ± 7 µm, P < .05). Crypt depth and tissue protein synthesis were higher in PN-COL (233 ± 7 µm, 22%/d) and PN-FORM (262 ± 13 µm, 22%/d) vs PN (190 ± 4 µm, 9%/d, both P < .05). In experiment 2, enteral colostrum supplementation was well tolerated, and no infants developed clinical signs of cow's milk allergy. CONCLUSION Minimal enteral nutrition feeding with bovine colostrum and formula induced similar intestinal adaptation after resection in piglets. Colostrum was well tolerated by newly resected infants, but the clinical indication for colostrum supplementation to infants subjected to intestinal resection remains to be determined.
Collapse
Affiliation(s)
- Lise Aunsholt
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Niels Qvist
- Department of Surgery, University Hospital of Odense, Odense, Denmark
| | - Per T Sangild
- Department of Veterinary Clinical and Animal Sciences, Frederiksberg C, Denmark.,Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen Ø, Denmark
| | - Andreas Vegge
- Department of Veterinary Clinical and Animal Sciences, Frederiksberg C, Denmark.,Global Research, Novo Nordisk, Måløv, Denmark
| | - Barbara Stoll
- Children's Nutrition Research Centre, Department of Paediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Douglas G Burrin
- Children's Nutrition Research Centre, Department of Paediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | - Thomas Eriksen
- Department of Veterinary Clinical and Animal Sciences, Frederiksberg C, Denmark
| | - Steffen Husby
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Thomas Thymann
- Department of Veterinary Clinical and Animal Sciences, Frederiksberg C, Denmark
| |
Collapse
|
31
|
Celi P, Cowieson A, Fru-Nji F, Steinert R, Kluenter AM, Verlhac V. Gastrointestinal functionality in animal nutrition and health: New opportunities for sustainable animal production. Anim Feed Sci Technol 2017. [DOI: 10.1016/j.anifeedsci.2017.09.012] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Rozé JC, Ancel PY, Lepage P, Martin-Marchand L, Al Nabhani Z, Delannoy J, Picaud JC, Lapillonne A, Aires J, Durox M, Darmaun D, Neu J, Butel MJ. Nutritional strategies and gut microbiota composition as risk factors for necrotizing enterocolitis in very-preterm infants. Am J Clin Nutr 2017; 106:821-830. [PMID: 28659297 PMCID: PMC5573022 DOI: 10.3945/ajcn.117.152967] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/05/2017] [Indexed: 12/22/2022] Open
Abstract
Background: The pathophysiology of necrotizing enterocolitis (NEC) remains poorly understood.Objective: We assessed the relation between feeding strategies, intestinal microbiota composition, and the development of NEC.Design: We performed a prospective nationwide population-based study, EPIPAGE 2 (Etude Epidémiologique sur les Petits Ages Gestationnels), including preterm infants born at <32 wk of gestation in France in 2011. From individual characteristics observed during the first week of life, we calculated a propensity score for the risk of NEC (Bell's stage 2 or 3) after day 7 of life. We analyzed the relation between neonatal intensive care unit (NICU) strategies concerning the rate of progression of enteral feeding, the direct-breastfeeding policy, and the onset of NEC using general linear mixed models to account for clustering by the NICU. An ancillary propensity-matched case-control study, EPIFLORE (Etude Epidémiologique de la flore), in 20 of the 64 NICUs, analyzed the intestinal microbiota by culture and 16S ribosomal RNA gene sequencing.Results: Among the 3161 enrolled preterm infants, 106 (3.4%; 95% CI: 2.8%, 4.0%) developed NEC. Individual characteristics were significantly associated with NEC. Slower and intermediate rates of progression of enteral feeding strategies were associated with a higher risk of NEC, with an adjusted OR of 2.3 (95% CI: 1.2, 4.5; P = 0.01) and 2.0 (95% CI: 1.1, 3.5; P = 0.02), respectively. Less favorable and intermediate direct-breastfeeding policies were associated with higher NEC risk as well, with an adjusted OR of 2.5 (95% CI: 1.1, 5.8; P = 0.03) and 2.3 (95% CI: 1.1, 4.8; P = 0.02), respectively. Microbiota analysis performed in 16 cases and 78 controls showed an association between Clostridium neonatale and Staphylococcus aureus with NEC (P = 0.001 and P = 0.002).Conclusions: A slow rate of progression of enteral feeding and a less favorable direct-breastfeeding policy are associated with an increased risk of developing NEC. For a given level of risk assessed by propensity score, colonization by C. neonatale and/or S. aureus is significantly associated with NEC. This trial (EPIFLORE study) was registered at clinicaltrials.gov as NCT01127698.
Collapse
Affiliation(s)
- Jean-Christophe Rozé
- Department of Neonatal Medicine, .,Epidémiologie Clinique, Clinical Investigation Center - Clinical Epidemiology (CIC004), and.,INRA, UMR 1280 Physiology of Nutritional Adaptations, Nantes University Hospital, Nantes, France
| | - Pierre-Yves Ancel
- INSERM, U1153, Obstetrical, Perinatal and Pediatric Epidemiology Team, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Paris Descartes University, Paris, France,Risks in Pregnancy Department, Paris Descartes University, Paris, France,Clinical investigation center CIC P1419, Cochin Hotel-Dieu Hospital, AP-HP, Paris, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Paris, France
| | - Laetitia Martin-Marchand
- INSERM, U1153, Obstetrical, Perinatal and Pediatric Epidemiology Team, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Paris Descartes University, Paris, France
| | - Ziad Al Nabhani
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Paris, France
| | - Johanne Delannoy
- Risks in Pregnancy Department, Paris Descartes University, Paris, France,EA 4065 Intestinal Ecosystem, Probiotics, Antibiotics, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | - Jean-Charles Picaud
- Department of Neonatal Medicine, Croix Rousse Hospital, Lyon Hospitals, Lyon, France
| | - Alexandre Lapillonne
- Department of Neonatal Medicine, AP-HP, Necker Enfants Malades Hospital, Paris, France
| | - Julio Aires
- Risks in Pregnancy Department, Paris Descartes University, Paris, France,EA 4065 Intestinal Ecosystem, Probiotics, Antibiotics, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | - Mélanie Durox
- INSERM, U1153, Obstetrical, Perinatal and Pediatric Epidemiology Team, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Paris Descartes University, Paris, France
| | - Dominique Darmaun
- INRA, UMR 1280 Physiology of Nutritional Adaptations, Nantes University Hospital, Nantes, France
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Marie-José Butel
- Risks in Pregnancy Department, Paris Descartes University, Paris, France,EA 4065 Intestinal Ecosystem, Probiotics, Antibiotics, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | | | | |
Collapse
|
33
|
Elsabagh M, Inabu Y, Obitsu T, Sugino T. Response of plasma glucagon-like peptide-2 to feeding pattern and intraruminal administration of volatile fatty acids in sheep. Domest Anim Endocrinol 2017; 60:31-41. [PMID: 28431319 DOI: 10.1016/j.domaniend.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 03/03/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide-2 (GLP-2), a gut peptide secreted by enteroendocrine L cells, has recently been identified as a key regulator of intestinal growth and absorptive function in ruminants. However, reports on GLP-2 secretion are few, and more information regarding its secretion dynamics is needed. In this study, two experiments were conducted to elucidate the daily rhythm of GLP-2 secretion in response to feeding regimen and to investigate the effect of volatile fatty acids (VFA) on GLP-2 release in sheep. In experiment 1, blood samples were collected over 3 d from 4 Suffolk mature wethers adapted to a maintenance diet fed once daily; day 1 sampling was preceded by 24 h of fasting to reach steady state. On days 1 and 3, samples were collected every 10 min from 11:00 to 14:00 on both days and then every 1 h until 00:00 on day 1 only; feed was offered at 12:00. On day 2, feed was withheld, and sampling was performed every hour from 01:00 to 00:00. In experiment 2, 5 Suffolk mature wethers were assigned to 5 treatment groups of intraruminal administration of saline, acetate, propionate, butyrate, or VFA mix (acetate, propionate, and butyrate in a ratio of 65:20:15) in a 5 × 5 Latin square design. Blood samples were collected at 0, 1.5, 3, 6, 9, 12, 15, 20, 25, 30, 40, 50, 60, 90, and 120 min relative to the beginning of administration at 12:00. In both experiments, plasma GLP-2, glucagon-like peptide-1 (GLP-1), glucose, insulin, and β-hydroxy butyric acid (BHBA) levels were measured. In experiment 1, incremental area under the curve was greater (P < 0.05) post-feeding than pre-feeding on days 1 and 3 for GLP-2 and tended to be greater (P < 0.1) on day 1 for GLP-1. Plasma insulin, glucose, and BHBA levels increased (P < 0.05) on day 1 post-feeding. Plasma GLP-2 was poorly correlated with GLP-1 but positively correlated with insulin, glucose, and BHBA. In experiment 2, administration of butyrate and VFA mix remarkably increased plasma GLP-2 (P = 0.05) and BHBA (P < 0.0001) levels compared with those in other treatments. Plasma GLP-1 levels were higher with butyrate administration compared with those in the saline, acetate, and VFA mix (P = 0.019). Propionate administration increased plasma glucose (P = 0.013) and insulin (P = 0.053) levels. Thus, our data confirmed that GLP-2 release is responsive to feeding and might be promoted by BHBA produced by the rumen epithelial metabolism of butyrate. Further molecular- and cellular-level studies are needed to determine the role of butyrate as a signaling molecule for GLP-2 release.
Collapse
Affiliation(s)
- M Elsabagh
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, 33516 Kafr El-Sheikh, Egypt
| | - Y Inabu
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - T Obitsu
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - T Sugino
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan.
| |
Collapse
|
34
|
Duodenal Disaccharidase Activities During and After Weaning off Parenteral Nutrition in Pediatric Intestinal Failure. J Pediatr Gastroenterol Nutr 2017; 64:777-782. [PMID: 27482764 DOI: 10.1097/mpg.0000000000001347] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Data on factors affecting absorptive function in children with intestinal failure (IF) are sparse. We evaluated duodenal disaccharidase activities and inflammation in relation to parenteral nutrition (PN) and intestinal resection in pediatric onset IF. METHODS Disaccharidase (maltase, sucrase, and lactase) activities and histologic inflammation were evaluated from duodenal biopsies in 58 patients during PN (n = 23) or full enteral nutrition (n = 40) and in 43 matched controls. The first and the last postresection biopsies were analyzed separately after 4.3 (1.2-9.7) years and 6.5 (2.3-12.4) years, respectively. RESULTS During PN, maltase and sucrase activities were 1.6-fold lower and mucosal inflammation more frequent (22% vs 3%) when compared to matched controls (P < 0.05 for both). In patients on full enteral nutrition, activities of maltase and sucrase were significantly higher than that in patients receiving PN and comparable to those of matched controls. Postresection time correlated positively (r = 0.448 and r = 0.369) and percentage length of the remaining small intestine inversely (r = -0.337 and r = -0.407) with maltase and sucrase activity in patients on full enteral nutrition (P < 0.05 for all), whereas proportional length of remaining colon correlated positively with maltase and lactase activity (r = 0.424-0.544, P < 0.05) in patients receiving PN. CONCLUSIONS In children with IF, PN dependency associated with decreased duodenal maltase and sucrase activities and mucosal inflammation, which may disturb intestinal absorptive function. Localization and extent of intestinal resection and post-resection time correlated with duodenal disaccharidase activities.
Collapse
|
35
|
Stoll B, Price PT, Reeds PJ, Chang X, Henry JF, van Goudoever JB, Holst JJ, Burrin DG. Feeding an Elemental Dietvsa Milk-Based Formula Does Not Decrease Intestinal Mucosal Growth in Infant Pigs. JPEN J Parenter Enteral Nutr 2017; 30:32-9. [PMID: 16387897 DOI: 10.1177/014860710603000132] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND We previously showed that the level of enteral nutrient intake determines the rate of intestinal growth in piglets. Our objective was to determine whether providing enteral nutrition in the form of elemental nutrients (glucose, amino acids, lipid [ED]) rather than cow's milk formula (lactose, protein, lipid [FORM]) reduces small intestinal growth and lactase activity. METHODS Three-week-old piglets were fed either ED (n = 7) intragastrically or FORM (n = 6) orally for 6 days. RESULTS Intestinal protein and DNA masses, villus height, and crypt depth were not different in ED and FORM pigs. Crypt cell proliferation, measured by in vivo bromodeoxyuridine labeling, was significantly (p < .05) higher (+37%) in ED than in FORM pigs. Rates of mucosal protein synthesis (%/d), measured by in vivo 2H-leucine incorporation, were higher (p < .05) in ED than FORM (147 vs 89) pigs. Circulating concentrations (pmol/L) of the intestinotrophic peptide, glucagon-like peptide-2 (GLP-2), were also higher (p < .05) in ED than in FORM (148 vs 87) pigs. The mean lactase-specific activity (micromol/min/g) in proximal and distal segments was higher (p < .05) in FORM than in ED (124 vs 58) pigs. CONCLUSIONS We conclude that intestinal mucosal growth and villus morphology are similar in pigs fed ED and FORM, despite higher cell proliferation and protein synthesis rates and lower lactase activity with ED. This implies that elemental diets may be as trophic as polymeric formulas to simultaneously provide nutrition and a stimulus for intestinal growth during bowel rest.
Collapse
Affiliation(s)
- Barbara Stoll
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Huang L, Ma XY, Jiang ZY, Hu YJ, Zheng CT, Yang XF, Wang L, Gao KG. Effects of soybean isoflavone on intestinal antioxidant capacity and cytokines in young piglets fed oxidized fish oil. J Zhejiang Univ Sci B 2016; 17:965-974. [PMID: 27921401 PMCID: PMC5172600 DOI: 10.1631/jzus.b1600078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/04/2016] [Indexed: 01/29/2023]
Abstract
To investigate the effect of glycitein, a synthetic soybean isoflavone (ISF), on the intestinal antioxidant capacity, morphology, and cytokine content in young piglets fed oxidized fish oil, 72 4-d-old male piglets were assigned to three treatments. The control group was fed a basal diet containing fresh fish oil, and the other two groups received the same diet except for the substitution with the same dosage of oxidized fish oil alone or with ISF (oxidized fish oil plus ISF). After 21 d of feeding, supplementation of oxidized fish oil increased the levels of malondialdehyde (MDA), oxidized glutathione (GSSG), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), nuclear factor κ B (NF-κB), inducible nitric oxide synthase (iNOS), NO, and Caspase-3 in jejunal mucosa, and decreased the villous height in duodenum and the levels of secretory immunoglobulin A (sIgA) and IL-4 in the jejunal mucosa compared with supplementation with fresh oil. The addition of oxidized fish oil plus ISF partially alleviated this negative effect. The addition of oxidized fish oil plus ISF increased the villous height and levels of sIgA and IL-4 in jejunal mucosa, but decreased the levels of IL-1β and IL-2 in jejunal mucosa (P<0.05) compared with oxidized fish oil. Collectively, these results show that dietary supplementation of ISF could partly alleviate the negative effect of oxidized fish oil by improving the intestinal morphology as well as the antioxidant capacity and immune function in young piglets.
Collapse
|
37
|
Yang H, Xiong X, Wang X, Li T, Yin Y. Effects of weaning on intestinal crypt epithelial cells in piglets. Sci Rep 2016; 6:36939. [PMID: 27830738 PMCID: PMC5103268 DOI: 10.1038/srep36939] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/11/2016] [Indexed: 01/08/2023] Open
Abstract
Intestinal epithelial cells in the crypt proliferate in piglets in response to weaning. However, the underlying mechanism has been unclear. We examined 40 piglets from eight litters (five piglets per litter) that were weaned at the age of 14 d, and one piglet from each litter was randomly selected for closer investigation. Based on the distended intestinal sac method, we isolated crypt epithelial cells from the mid-jejunum on Days 0, 1, 3, 5, and 7 post-weaning. Protein expression was analyzed using either isobaric tags for relative and absolute quantification or western blotting. Proteins related to the cell cycle, organization of the cellular macromolecular complex subunit, localization of cellular macromolecules, Golgi vesicle transport, fatty acid metabolism, oxidative phosphorylation, and translational initiation were mainly down-regulated, while those involved in glycolysis, cell cycle arrest, protein catabolism, and cellular amino acid metabolism were up-regulated. The amount of proteins active in the mTOR signaling pathway was generally decreased over time. These results indicate that weaning influences energy metabolism, cellular macromolecule organization and localization, and protein metabolism, thereby affecting the proliferation of intestinal epithelial cells in weaned piglets. Moreover, those cellular processes are possibly controlled by that signaling pathway.
Collapse
Affiliation(s)
- Huansheng Yang
- Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, China.,Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Xia Xiong
- Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Xiaocheng Wang
- Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Tiejun Li
- Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yulong Yin
- Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, China.,Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| |
Collapse
|
38
|
de Oliveira DC, da Silva Lima F, Sartori T, Santos ACA, Rogero MM, Fock RA. Glutamine metabolism and its effects on immune response: molecular mechanism and gene expression. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s41110-016-0016-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
39
|
Lim DW, Diané A, Muto M, Vine DF, Nation PN, Wizzard PR, Sigalet DL, Bigam DL, Pencharz PB, Turner JM, Wales PW. Differential Effects on Intestinal Adaptation Following Exogenous Glucagon-Like Peptide 2 Therapy With and Without Enteral Nutrition in Neonatal Short Bowel Syndrome. JPEN J Parenter Enteral Nutr 2016; 41:156-170. [DOI: 10.1177/0148607116665812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- David W. Lim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Abdoulaye Diané
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Mitsuru Muto
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Donna F. Vine
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick N. Nation
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela R. Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - David L. Sigalet
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - David L. Bigam
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Paul B. Pencharz
- Department of Nutritional Sciences and Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Justine M. Turner
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Paul W. Wales
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, Hospital for Sick Children & University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Wu C, Wang X, Jiang T, Li C, Zhang L, Gao X, Tian F, Li N, Li J. Partial Enteral Nutrition Mitigated Ischemia/Reperfusion-Induced Damage of Rat Small Intestinal Barrier. Nutrients 2016; 8:nu8080502. [PMID: 27548209 PMCID: PMC4997415 DOI: 10.3390/nu8080502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/07/2016] [Accepted: 08/11/2016] [Indexed: 01/03/2023] Open
Abstract
Background and Aims: This study was designed to investigate a relatively optimum dose of partial enteral nutrition (PEN) which effectively attenuates intestinal barrier dysfunction initiated by ischemia/reperfusion injury (IRI). Methods: In experiment 1, 60 male Sprague-Dawley (SD) rats were subjected to intestinal IRI and assigned to six groups according to the different proportion of EN administrations: namely total parenteral nutrition (TPN or 0%EN), 10%EN, 20%EN, 40%EN, 60%EN, and total enteral nutrition (TEN or 100%) groups, the deficits of intraluminal calorie were supplemented by PN. In experiment 2, 50 male SD rats were subjected to intestinal IRI and divided into five groups based on the results of experiment 1: TPN, TEN, 20%EN, TPN plus pretreatment with NF-κB antagonist 30 min before IRI (TPN+PDTC), and TPN plus pretreatment with HIF-1α antagonist 30 min before IRI (TPN+YC-1) groups. Results: In experiment 1, previous IRI combined with subsequent EN shortage disrupted the structure of intestinal epithelial cell and tight junctions (TJs). While 20% dose of EN had an obviously protective effect on these detrimental consequences. In experiment 2, compared with TPN only, 20%EN exerted a significant protection of barrier function of intestinal epithelium. Analogous results were observed when TPN combined with specific NF-κB/HIF-1α inhibitors (PDTC and YC-1). Meanwhile, the expression of NF-κB/HIF-1α had a similar trend among the groups. Conclusions: Our findings indicate that 20%EN is the minimally effective dosage of EN which promotes the recovery of intestinal barrier function after IRI in a rat model. Furthermore, we discreetly speculate that this benefit is, at least partly, related to NF-κB/HIF-1α pathway expression.
Collapse
Affiliation(s)
- Chao Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xinying Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Tingting Jiang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Chaojun Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University and Model Animal Research Center, National Resource Center for Mutant Mice, Nanjing 210093, China.
| | - Li Zhang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Xuejin Gao
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Feng Tian
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Ning Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| |
Collapse
|
41
|
Thymann T. Endocrine regulation of gut maturation in early life in pigs. Domest Anim Endocrinol 2016; 56 Suppl:S90-3. [PMID: 27345327 DOI: 10.1016/j.domaniend.2016.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 01/17/2023]
Abstract
After birth, the newborn must adapt to the acute challenges of circulatory changes, active respiration, thermoregulation, microbial colonization, and enteral nutrition. Whereas these processes normally occur without clinical complications in neonates born at term, birth at a preterm state of gestation is associated with high morbidity and mortality. In commercial pig production, perinatal mortality is higher than in any other mammalian species. Asphyxia, hypothermia, hypoglycemia, sepsis, and gut dysmotility, represent some of the most common findings. The intestine is a particularly sensitive organ after birth, as it must adapt acutely to enteral nutrition and microbial colonization. Likewise, during the weaning phase, the intestine must adapt to new diet types. Both critical phases are associated with high morbidity. This review focuses on the endocrine changes occurring around birth and weaning. There are a number of endocrine adaptations in late gestation and early postnatal life that are under influence of development stage and environmental factors such as diet. The review discusses general endocrine changes in perinatal life but specifically focuses on the role of glucagon-like peptide-2. This gut-derived hormone plays a key role in development and function of the intestine in early life.
Collapse
Affiliation(s)
- T Thymann
- Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, Department of Clinical Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, DK-1870, Denmark.
| |
Collapse
|
42
|
Castro J, Morrison S, Hosseinni A, Loor J, Drackley J, Ipharraguerre I. Secretion of glucagon-like peptide-2 responds to nutrient intake but not glucose provision in milk-fed calves. J Dairy Sci 2016; 99:5793-5807. [DOI: 10.3168/jds.2015-10519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/20/2016] [Indexed: 11/19/2022]
|
43
|
Nutrient-intake-level-dependent regulation of intestinal development in newborn intrauterine growth-restricted piglets via glucagon-like peptide-2. Animal 2016; 10:1645-54. [PMID: 27095347 DOI: 10.1017/s1751731116000690] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The objective of the present study was to investigate the intestinal development of newborn intrauterine growth-restricted (IUGR) piglets subjected to normal nutrient intake (NNI) or restricted nutrient intake (RNI). Newborn normal birth weight (NBW) and IUGR piglets were allotted to NNI or RNI levels for 4 weeks from day 8 postnatal. IUGR piglets receiving NNI had similar growth performance compared with that of NBW piglets. Small intestine length and villous height were greater in IUGR piglets fed the NNI than that of piglets fed the RNI. Lactase activity was increased in piglets fed the NNI compared with piglets fed the RNI. Absorptive function, represented by active glucose transport by the Ussing chamber method and messenger RNA (mRNA) expressions of two main intestinal glucose transporters, Na+-dependent glucose transporter 1 (SGLT1) and glucose transporter 2 (GLUT2), were greater in IUGR piglets fed the NNI compared with piglets fed the RNI regimen. The apoptotic process, characterized by caspase-3 activity (a sign of activated apoptotic cells) and mRNA expressions of p53 (pro-apoptotic), bcl-2-like protein 4 (Bax) (pro-apoptotic) and B-cell lymphoma-2 (Bcl-2) (anti-apoptotic), were improved in IUGR piglets fed the NNI regimen. To test the hypothesis that improvements in intestinal development of IUGR piglets fed NNI might be mediated through circulating glucagon-like peptide-2 (GLP-2), GLP-2 was injected subcutaneously to IUGR piglets fed the RNI from day 8 to day 15 postnatal. Although the intestinal development of IUGR piglets fed the RNI regimen was suppressed compared with those fed the NNI regimen, an exogenous injection of GLP-2 was able to bring intestinal development to similar levels as NNI-fed IUGR piglets. Collectively, our results demonstrate that IUGR neonates that have NNI levels could improve intestinal function via the regulation of GLP-2.
Collapse
|
44
|
Hansen CF, Thymann T, Andersen AD, Holst JJ, Hartmann B, Hilsted L, Langhorn L, Jelsing J, Sangild PT. Rapid gut growth but persistent delay in digestive function in the postnatal period of preterm pigs. Am J Physiol Gastrointest Liver Physiol 2016; 310:G550-60. [PMID: 26822913 PMCID: PMC4836131 DOI: 10.1152/ajpgi.00221.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/22/2016] [Indexed: 01/31/2023]
Abstract
Preterm infants often tolerate full enteral nutrition a few weeks after birth but it is not known how this is related to gut maturation. Using pigs as models, we hypothesized that intestinal structure and digestive function are similar in preterm and term individuals at 3-4 wk after birth and that early enteral nutrition promotes maturation. Preterm or term cesarean-delivered pigs were fed total parenteral nutrition, or partial enteral nutrition [Enteral (Ent), 16-64 ml·kg(-1)·day(-1) of bovine colostrum] for 5 days, followed by full enteral milk feeding until day 26 The intestine was collected for histological and biochemical analyses at days 0, 5, and 26 (n = 8-12 in each of 10 treatment groups). Intestinal weight (relative to body weight) was reduced in preterm pigs at 0-5 days but ENT feeding stimulated the mucosal volume and peptidase activities. Relative to term pigs, mucosal volume remained reduced in preterm pigs until 26 days although plasma glucagon-like peptide 2 (GLP-2) and glucose-dependent insulin-trophic peptide (GIP) levels were increased. Preterm pigs also showed reduced hexose absorptive capacity and brush-border enzyme (sucrase, maltase) activities at 26 days, relative to term pigs. Intestinal structure shows a remarkable growth adaptation in the first week after preterm birth, especially with enteral nutrition, whereas some digestive functions remain immature until at least 3-4 wk. It is important to identify feeding regimens that stimulate intestinal maturation in the postnatal period of preterm infants because some intestinal functions may show long-term developmental delay.
Collapse
Affiliation(s)
- Carl Frederik Hansen
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | - Thomas Thymann
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | | | - Jens Juul Holst
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Bolette Hartmann
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Linda Hilsted
- 4Department of Clinical Biochemistry, Copenhagen University Hospital, Cophenhagen Denmark; and
| | - Louise Langhorn
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | | | - Per Torp Sangild
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark; ,5Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Denmark
| |
Collapse
|
45
|
Yang C, Yang X, Lackeyram D, Rideout TC, Wang Z, Stoll B, Yin Y, Burrin DG, Fan MZ. Expression of apical Na(+)-L-glutamine co-transport activity, B(0)-system neutral amino acid co-transporter (B(0)AT1) and angiotensin-converting enzyme 2 along the jejunal crypt-villus axis in young pigs fed a liquid formula. Amino Acids 2016; 48:1491-508. [PMID: 26984322 DOI: 10.1007/s00726-016-2210-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/29/2016] [Indexed: 01/11/2023]
Abstract
Gut apical amino acid (AA) transport activity is high at birth and during suckling, thus being essential to maintain luminal nutrient-dependent mucosal growth through providing AA as essential metabolic fuel, substrates and nutrient stimuli for cellular growth. Because system-B(0) Na(+)-neutral AA co-transporter (B(0)AT1, encoded by the SLC6A19 gene) plays a dominant role for apical uptake of large neutral AA including L-Gln, we hypothesized that high apical Na(+)-Gln co-transport activity, and B(0)AT1 (SLC6A19) in co-expression with angiotensin-converting enzyme 2 (ACE2) were expressed along the entire small intestinal crypt-villus axis in young animals via unique control mechanisms. Kinetics of Na(+)-Gln co-transport activity in the apical membrane vesicles, prepared from epithelial cells sequentially isolated along the jejunal crypt-villus axis from liquid formula-fed young pigs, were measured with the membrane potential being clamped to zero using thiocyanate. Apical maximal Na(+)-Gln co-transport activity was much higher (p < 0.05) in the upper villus cells than in the middle villus (by 29 %) and the crypt (by 30 %) cells, whereas Na(+)-Gln co-transport affinity was lower (p < 0.05) in the upper villus cells than in the middle villus and the crypt cells. The B(0)AT1 (SLC6A19) mRNA abundance was lower (p < 0.05) in the crypt (by 40-47 %) than in the villus cells. There were no significant differences in B(0)AT1 and ACE2 protein abundances on the apical membrane among the upper villus, the middle villus and the crypt cells. Our study suggests that piglet fast growth is associated with very high intestinal apical Na(+)-neutral AA uptake activities via abundantly co-expressing B(0)AT1 and ACE2 proteins in the apical membrane and by transcribing the B(0)AT1 (SLC6A19) gene in the epithelia along the entire crypt-villus axis.
Collapse
Affiliation(s)
- Chengbo Yang
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada. .,Department of Animal Science, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Xiaojian Yang
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.,Southern Research and Outreach Center, University of Minnesota, Waseca, MN, 56093, USA
| | - Dale Lackeyram
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Todd C Rideout
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.,Department of Exercise and Nutrition Sciences, the State University of New York at Buffalo, New York, 14214, USA
| | - Zirong Wang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, Xinjiang, China
| | - Barbara Stoll
- US Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yulong Yin
- Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Douglas G Burrin
- US Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ming Z Fan
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
46
|
Lim DW, Wales PW, Turner JM, Bigam DL, Brubaker PL. On the horizon: trophic peptide growth factors as therapy for neonatal short bowel syndrome. Expert Opin Ther Targets 2016; 20:819-30. [DOI: 10.1517/14728222.2016.1146695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- David W. Lim
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Paul W. Wales
- Department of Surgery, University of Toronto & Hospital for Sick Children, Toronto, ON, Canada
| | - Justine M. Turner
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - David L. Bigam
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Patricia L. Brubaker
- Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
47
|
Xiao Y, Yan W, Lu L, Wang Y, Lu W, Cao Y, Cai W. p38/p53/miR-200a-3p feedback loop promotes oxidative stress-mediated liver cell death. Cell Cycle 2016; 14:1548-58. [PMID: 25789565 DOI: 10.1080/15384101.2015.1026491] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although our previous studies have provided evidence that oxidative stress has an essential role in total parenteral nutrition (TPN)-associated liver injury, the mechanisms involved are incompletely understood. Here, we show the existence of crosstalk between the miR-200 family of microRNAs and oxidative stress. The members of the miR-200 family are markedly enhanced in hepatic cells by hydrogen peroxide (H2O2) treatment. The upregulation of miR-200-3p in turn modulates the H2O2-mediated oxidative stress response by targeting p38α. The enhanced expression of miR-200-3p mimics p38α deficiency and promotes H2O2-induced cell death. Members of the miR-200 family that are known to inhibit the epithelial to mesenchymal transition (EMT) are induced by the tumor suppressor p53. Here, we show that p53 phosphorylation at Ser 33 contributes to H2O2-induced miR-200s transcription. In addition, we show that p38α can directly phosphorylate p53 at serine 33 upon H2O2 exposure. Thus, we suggest that in liver cells, the oxidative stress-induced, p38α-mediated phosphorylation of p53 at Ser33 is essential for the functional regulation of oxidative stress-induced miR-200 transcription by p53. Collectively, our data indicate that the p53-dependent expression of miR-200a-3p promotes cell death by inhibiting a p38/p53/miR-200 feedback loop.
Collapse
Affiliation(s)
- Yongtao Xiao
- a Department of Pediatric Surgery ; Xinhua Hospital; School of Medicine; Shanghai Jiao Tong University ; Shanghai , China
| | | | | | | | | | | | | |
Collapse
|
48
|
Akotia DH, Durham JT, Arnell KM, Petruzzelli DL, Katheria AC. Relationship Between Near-Infrared Spectroscopy and Transabdominal Ultrasonography: Noninvasive Monitoring of Intestinal Function in Neonates. Med Sci Monit 2016; 22:61-8. [PMID: 26736134 PMCID: PMC4708098 DOI: 10.12659/msm.895730] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Near-infrared spectroscopy (NIRS) has the potential to continuously and noninvasively monitor intestinal function. This technology may be valuable because among neonates, intestinal maturity is highly variable and difficult to assess based solely on clinical signs. The aim of this study was to determine if there is an association between NIRS-based StO2 measurements and peristaltic activity assessed by transabdominal ultrasonography (US). MATERIAL/METHODS Nineteen neonates of gestational age >32 weeks were categorized according to "no/low" versus "normal/hyperactive" motility levels, based on blinded US scan results. StO2 was recorded every 2 s for 24 h, following the ultrasound recording. Differences between the resulting estimates of average StO2 (bias of fits) and goodness-of-fit (residuals) were evaluated. RESULTS Newborns with normal/hyperactive motility had higher mean StO2 than newborns with no/low motility (72.3±4.4 vs. 65.5±7.9, p<0.05, F=5.65). Residual errors were not significantly different between the 2 groups (p=0.213, F=0.213). A multivariate linear regression model using the means, residuals, and pairwise products of both, demonstrated more significant separation (0.47±0.26 vs. -0.24±0.33, p<0.01, F=27.4). A non-linear variant of the multivariate linear regression model demonstrated greatest separation (0.68±0.24 vs. -0.49±0.53, p<0.01, F=41.9). CONCLUSIONS This is the first study to demonstrate an association between NIRS-based StO2 measurements and peristaltic activity visualized by ultrasound imaging. NIRS may offer a continuous, noninvasive method to assess motility. This may have significant implications in premature infants at risk for feeding intolerance or necrotizing enterocolitis.
Collapse
Affiliation(s)
- Devang H Akotia
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, CA, USA
| | - Jayson T Durham
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, CA, USA
| | - Kathy M Arnell
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, CA, USA
| | - Deborah L Petruzzelli
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, CA, USA
| | - Anup C Katheria
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, CA, USA
| |
Collapse
|
49
|
Abstract
OBJECTIVE We aimed to review gastric dysmotility in critically ill children: 1) its pathophysiology, with a focus on critical care diseases and therapies that affect gastric motility, 2) diagnostic methodologies, and 3) current and future potential therapies. DATA SOURCES Eligible studies were identified from PubMed and MEDLINE. STUDY SELECTION Literature search included the following key terms: "gastric emptying," "gastric motility/dysmotility," "gastrointestinal motility/dysmotility," "nutrition intolerance," and "gastric residual volume." DATA EXTRACTION Studies since 1995 were extracted and reviewed for inclusion by the authors related to the physiology, pathophysiology, diagnostic methodologies, and available therapies for gastric emptying. DATA SYNTHESIS Delayed gastric emptying, a common presentation of gastric dysmotility, is present in up to 50% of critically ill children. It is associated with the potential for aspiration, ventilator-associated pneumonia, and inadequate delivery of enteral nutrition and may affect the efficacy of enteral medications, all of which may be result in poor patient outcomes. Gastric motility is affected by critical illness and its associated therapies. Currently available diagnostic tools to identify gastric emptying at the bedside have not been systematically studied and applied in this cohort. Gastric residual volume measurement, used as an indirect marker of delayed gastric emptying in PICUs around the world, may be inaccurate. CONCLUSIONS Gastric dysmotility is common in critically ill children and impacts patient safety and outcomes. However, it is poorly understood, inadequately defined, and current therapies are limited and based on scant evidence. Understanding gastric motility and developing accurate bedside measures and novel therapies for gastric emptying are highly desirable and need to be further investigated.
Collapse
|
50
|
Kuiken NSS, Rings EHHM, Havinga R, Groen AK, Tissing WJE. Effect of minimal enteral feeding on recovery in a methotrexate-induced gastrointestinal mucositis rat model. Support Care Cancer 2015; 24:1357-64. [PMID: 26335404 PMCID: PMC4729808 DOI: 10.1007/s00520-015-2911-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/16/2015] [Indexed: 11/25/2022]
Abstract
PURPOSE Patients suffering from gastrointestinal mucositis often receive parenteral nutrition as nutritional support. However, the absence of enteral nutrition might not be beneficial for the intestine. We aimed to determine the feasibility of minimal enteral feeding (MEF) administration in a methotrexate (MTX)-induced mucositis rat model and thereby determine the effect of MEF on recovery. METHODS Male Wistar rats were attached to swivel systems from day 1 to 5 after 45 mg/kg MTX IV injection. The MTX group continued ad libitum feeding, and the MTX + MEF group continued ad libitum feeding and received from day 1 to 5 continuously MEF. MEF consisted of 20% of their normal caloric intake. We measured body weight, intake, and plasma citrulline. At day 10, the rats were terminated and villus and crypt length were measured. RESULTS The administration of MEF caused no increased severity of mucositis phenotype, with comparable caloric intake, body weight, and plasma citrulline during mucositis. The recovery of plasma citrulline levels was not different between both groups. At day 7 and 8, the MTX + MEF group gained significantly more weight (p < 0.05 and p < 0.01, respectively), and at day 8 and 9 the total caloric intake was significantly increased (p < 0.01 and p < 0.05, respectively) compared to the MTX group. At day 10, the rats from the MTX + MEF group showed a significant increase in jejunal villus length compared to the MTX group (p < 0.05). CONCLUSIONS This is the first study in which the feasibility of MEF administration during chemotherapy-induced mucositis was determined. This study indicates that MEF administration is feasible during mucositis and suggests that MEF accelerates recovery after MTX-induced mucositis.
Collapse
Affiliation(s)
- Nicoline S S Kuiken
- Department of Pediatric Oncology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pediatric Gastroenterology and Hepatology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Edmond H H M Rings
- Department of Pediatrics, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
- Department of Pediatrics, Erasmus Medical Center Rotterdam, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Rick Havinga
- Department of Pediatric Gastroenterology and Hepatology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Albert K Groen
- Department of Pediatric Gastroenterology and Hepatology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Wim J E Tissing
- Department of Pediatric Oncology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|