1
|
Swanson K, Bell J, Hendrix D, Jiang D, Kutzler M, Batty B, Hanlon M, Bionaz M. Bovine milk consumption affects the transcriptome of porcine adipose stem cells: Do exosomes play any role? PLoS One 2024; 19:e0302702. [PMID: 39705291 DOI: 10.1371/journal.pone.0302702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/03/2024] [Indexed: 12/22/2024] Open
Abstract
The potential association of milk with childhood obesity has been widely debated and researched. Milk is known to contain many bioactive compounds as well as bovine exosomes rich in micro-RNA (miR) that can have effects on various cells, including stem cells. Among them, adipose stem cells (ASC) are particularly interesting due to their role in adipose tissue growth and, thus, obesity. The objective of this study was to evaluate the effect of milk consumption on miR present in circulating exosomes and the transcriptome of ASC in piglets. Piglets were supplemented for 11 weeks with 750 mL of whole milk (n = 6; M) or an isocaloric maltodextrin solution (n = 6; C). After euthanasia, ASC were isolated, quantified, and characterized. RNA was extracted from passage 1 ASC and sequenced. Exosomes were isolated and quantified from the milk and plasma of the pigs at 6-8 hours after milk consumption, and miRs were isolated from exosomes and sequenced. The transfer of exosomes from milk to porcine plasma was assessed by measuring bovine milk-specific miRs and mRNA in exosomes isolated from the plasma of 3 piglets during the first 6h after milk consumption. We observed a higher proportion of exosomes in the 80 nM diameter, enriched in milk, in M vs. C pigs. Over 500 genes were differentially expressed (DEG) in ASC isolated from M vs. C pigs. Bioinformatic analysis of DEG indicated an inhibition of the immune, neuronal, and endocrine systems and insulin-related pathways in ASC of milk-fed pigs compared with maltodextrin-fed pigs. Of the 900 identified miRs in porcine plasma exosomes, only 3 miRs were differentially abundant between the two groups and could target genes associated with neuronal functions. We could not detect exosomal miRs or mRNA transfer from milk to porcine-circulating plasma exosomes. Our data highlights the significant nutrigenomic role of milk consumption on ASC, a finding that does not appear to be attributed to miRs in bovine milk exosomes. The downregulation of insulin resistance and inflammatory-related pathways in the ASC of milk-fed pigs should be further explored in relation to milk and human health. In conclusion, the bioinformatic analyses and the absence of bovine exosomal miRs in porcine plasma suggest that miRs are not vertically transferred from milk exosomes.
Collapse
Affiliation(s)
- Katherine Swanson
- Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Jimmy Bell
- Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, United States of America
| | - David Hendrix
- Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, United States of America
| | - Duo Jiang
- Statistics, Oregon State University, Corvallis, Oregon, United States of America
| | - Michelle Kutzler
- Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Brandon Batty
- Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Melanie Hanlon
- Food Science and Technology, Oregon State University, Corvallis, Oregon, United States of America
| | - Massimo Bionaz
- Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
2
|
Weil PP, Reincke S, Hirsch CA, Giachero F, Aydin M, Scholz J, Jönsson F, Hagedorn C, Nguyen DN, Thymann T, Pembaur A, Orth V, Wünsche V, Jiang PP, Wirth S, Jenke ACW, Sangild PT, Kreppel F, Postberg J. Uncovering the gastrointestinal passage, intestinal epithelial cellular uptake and AGO2 loading of milk miRNAs in neonates using xenomiRs as tracers. Am J Clin Nutr 2023:S0002-9165(23)46299-5. [PMID: 36963568 DOI: 10.1016/j.ajcnut.2023.03.016] [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: 10/19/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND Human breast milk has a high microRNA (miRNA) content. It remains unknown whether and how milk miRNAs might affect intestinal gene regulation and homeostasis of the developing microbiome after initiation of enteral nutrition. However, this requires that relevant milk miRNA amounts survive gastrointestinal passage, are taken up by cells, and become available to the RNA interference (RNAi) machinery. It seems important to dissect the fate of these miRNAs after oral ingestion and gastrointestinal passage. OBJECTIVE Our goal was to analyze the potential transmissibility of milk miRNAs via the gastrointestinal system in neonate humans and a porcine model in vivo to contribute to the discussion whether milk miRNAs could influence gene regulation in neonates and thus might vertically transmit developmental relevant signals. DESIGN We performed cross-species profiling of miRNAs via deep-sequencing and utilized dietary xenobiotic taxon-specific milk miRNA (xenomiRs) as tracers in human and porcine neonates, followed by functional studies in primary human fetal intestinal epithelial cells (HIEC-6) using Ad5-mediated miRNA-gene transfer. RESULTS Mammals share many milk miRNAs yet exhibit taxon-specific miRNA fingerprints. We traced bovine-specific miRNAs from formula-nutrition in human preterm stool and 9 days after onset of enteral feeding in intestinal cells of preterm piglets. Thereafter, several xenomiRs accumulated in the intestinal cells. Moreover, few hours after introducing enteral feeding in preterm piglets with supplemented reporter miRNAs (cel-miR-39-5p/-3p), we observed their enrichment in blood serum and in AGO2-immunocomplexes from intestinal biopsies. CONCLUSIONS Milk-derived miRNAs survived gastrointestinal passage in human and porcine neonates. Bovine-specific miRNAs accumulated in intestinal cells of preterm piglets after enteral feeding with bovine colostrum/formula. In piglets, colostrum supplementation with cel-miR-39-5p/-3p resulted in increased blood levels of cel-miR-39-3p and argonaute RISC catalytic component 2 (AGO2) loading in intestinal cells. This suggests the possibility of vertical transmission of miRNA signaling from milk through the neonatal digestive tract.
Collapse
Affiliation(s)
- Patrick Philipp Weil
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| | - Susanna Reincke
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| | - Christian Alexander Hirsch
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| | - Federica Giachero
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| | - Malik Aydin
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany; HELIOS University Hospital Wuppertal, Children's Hospital, Centre for Clinical & Translational Research (CCTR), Witten/Herdecke University, Heusnerstr. 40, 42283 Wuppertal, Germany.
| | - Jonas Scholz
- Chair of Biochemistry and Molecular Medicine, Faculty of Health, Centre for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany.
| | - Franziska Jönsson
- Chair of Biochemistry and Molecular Medicine, Faculty of Health, Centre for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany.
| | - Claudia Hagedorn
- Chair of Biochemistry and Molecular Medicine, Faculty of Health, Centre for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany.
| | - Duc Ninh Nguyen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anton Pembaur
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| | - Valerie Orth
- HELIOS University Hospital Wuppertal, Department of Surgery II, Centre for Clinical & Translational Research (CCTR), Witten/Herdecke University, Heusnerstr. 40, 42283 Wuppertal, Germany.
| | - Victoria Wünsche
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| | - Ping-Ping Jiang
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark; School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Stefan Wirth
- HELIOS University Hospital Wuppertal, Children's Hospital, Centre for Clinical & Translational Research (CCTR), Witten/Herdecke University, Heusnerstr. 40, 42283 Wuppertal, Germany.
| | - Andreas C W Jenke
- Klinikum Kassel, Zentrum für Kinder- und Jugendmedizin, Neonatologie und allgemeine Pädiatrie, Mönchebergstr. 41-43, 34125 Kassel, Germany.
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Florian Kreppel
- Chair of Biochemistry and Molecular Medicine, Faculty of Health, Centre for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany.
| | - Jan Postberg
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany.
| |
Collapse
|
3
|
Embleton ND, Jennifer Moltu S, Lapillonne A, van den Akker CHP, Carnielli V, Fusch C, Gerasimidis K, van Goudoever JB, Haiden N, Iacobelli S, Johnson MJ, Meyer S, Mihatsch W, de Pipaon MS, Rigo J, Zachariassen G, Bronsky J, Indrio F, Köglmeier J, de Koning B, Norsa L, Verduci E, Domellöf M. Enteral Nutrition in Preterm Infants (2022): A Position Paper From the ESPGHAN Committee on Nutrition and Invited Experts. J Pediatr Gastroenterol Nutr 2023; 76:248-268. [PMID: 36705703 DOI: 10.1097/mpg.0000000000003642] [Citation(s) in RCA: 116] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES To review the current literature and develop consensus conclusions and recommendations on nutrient intakes and nutritional practice in preterm infants with birthweight <1800 g. METHODS The European Society of Pediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) Committee of Nutrition (CoN) led a process that included CoN members and invited experts. Invited experts with specific expertise were chosen to represent as broad a geographical spread as possible. A list of topics was developed, and individual leads were assigned to topics along with other members, who reviewed the current literature. A single face-to-face meeting was held in February 2020. Provisional conclusions and recommendations were developed between 2020 and 2021, and these were voted on electronically by all members of the working group between 2021 and 2022. Where >90% consensus was not achieved, online discussion meetings were held, along with further voting until agreement was reached. RESULTS In general, there is a lack of strong evidence for most nutrients and topics. The summary paper is supported by additional supplementary digital content that provide a fuller explanation of the literature and relevant physiology: introduction and overview; human milk reference data; intakes of water, protein, energy, lipid, carbohydrate, electrolytes, minerals, trace elements, water soluble vitamins, and fat soluble vitamins; feeding mode including mineral enteral feeding, feed advancement, management of gastric residuals, gastric tube placement and bolus or continuous feeding; growth; breastmilk buccal colostrum, donor human milk, and risks of cytomegalovirus infection; hydrolyzed protein and osmolality; supplemental bionutrients; and use of breastmilk fortifier. CONCLUSIONS We provide updated ESPGHAN CoN consensus-based conclusions and recommendations on nutrient intakes and nutritional management for preterm infants.
Collapse
Affiliation(s)
| | | | | | - Chris H P van den Akker
- the Department of Pediatrics - Neonatology, Amsterdam UMC - Emma Children's Hospital, University of Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Virgilio Carnielli
- Polytechnic University of Marche and Division of Neonatology, Ospedali Riuniti, Ancona, Ancona, Italy
| | - Christoph Fusch
- the Department of Pediatrics, Nuremberg General Hospital, Paracelsus Medical School, Nuremberg, Germany
- the Division of Neonatology, Department of Pediatrics, Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Konstantinos Gerasimidis
- the Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Johannes B van Goudoever
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Nadja Haiden
- the Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Silvia Iacobelli
- the Réanimation Néonatale et Pédiatrique, Néonatologie - CHU La Réunion, Saint-Pierre, France
| | - Mark J Johnson
- the Department of Neonatal Medicine, University Hospital Southampton NHS Trust, Southampton, UK
- the National Institute for Health Research Biomedical Research Centre Southampton, University Hospital Southampton NHS Trust and University of Southampton, Southampton, UK
| | - Sascha Meyer
- the Department of General Paediatrics and Neonatology, University Hospital of Saarland, Homburg, Germany
| | - Walter Mihatsch
- the Department of Pediatrics, Ulm University, Ulm, Germany
- the Department of Health Management, Neu-Ulm University of Applied Sciences, Neu-Ulm, Germany
| | - Miguel Saenz de Pipaon
- the Department of Pediatrics-Neonatology, La Paz University Hospital, Autonoma University of Madrid, Madrid, Spain
| | - Jacques Rigo
- the Neonatal Unit, University of Liège, CHR Citadelle, Liège, Belgium
| | - Gitte Zachariassen
- H.C. Andersen Children's Hospital, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Jiri Bronsky
- the Department of Paediatrics, University Hospital Motol, Prague, Czech Republic
| | - Flavia Indrio
- the Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Jutta Köglmeier
- the Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Barbara de Koning
- the Paediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lorenzo Norsa
- the Paediatric Hepatology, Gastroenterology and Transplantation, ASST Papa Giovanni XXIIII, Bergamo, Italy
| | - Elvira Verduci
- the Department of Health Sciences, University of Milan, Milan, Italy
- the Department of Paediatrics, Ospedale dei Bambini Vittore Buzzi, Milan, Italy
| | - Magnus Domellöf
- the Department of Clinical Sciences, Paediatrics, Umeå University, Umeå, Sweden
| |
Collapse
|
4
|
Mank E, Sáenz de Pipaón M, Lapillonne A, Carnielli VP, Senterre T, Shamir R, van Toledo L, van Goudoever JB. Efficacy and Safety of Enteral Recombinant Human Insulin in Preterm Infants: A Randomized Clinical Trial. JAMA Pediatr 2022; 176:452-460. [PMID: 35226099 PMCID: PMC8886453 DOI: 10.1001/jamapediatrics.2022.0020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Feeding intolerance is a common condition among preterm infants owing to immaturity of the gastrointestinal tract. Enteral insulin appears to promote intestinal maturation. The insulin concentration in human milk declines rapidly post partum and insulin is absent in formula; therefore, recombinant human (rh) insulin for enteral administration as a supplement to human milk and formula may reduce feeding intolerance in preterm infants. OBJECTIVE To assess the efficacy and safety of 2 different dosages of rh insulin as a supplement to both human milk and preterm formula. DESIGN, SETTING, AND PARTICIPANTS The FIT-04 multicenter, double-blind, placebo-controlled randomized clinical trial was conducted at 46 neonatal intensive care units throughout Europe, Israel, and the US. Preterm infants with a gestational age (GA) of 26 to 32 weeks and a birth weight of 500 g or more were enrolled between October 9, 2016, and April 25, 2018. Data were analyzed in January 2020. INTERVENTIONS Preterm infants were randomly assigned to receive low-dose rh insulin (400-μIU/mL milk), high-dose rh insulin (2000-μIU/mL milk), or placebo for 28 days. MAIN OUTCOMES AND MEASURES The primary outcome was time to achieve full enteral feeding (FEF) defined as an enteral intake of 150 mL/kg per day or more for 3 consecutive days. RESULTS The final intention-to-treat analysis included 303 preterm infants (low-dose group: median [IQR] GA, 29.1 [28.1-30.4] weeks; 65 boys [59%]; median [IQR] birth weight, 1200 [976-1425] g; high-dose group: median [IQR] GA, 29.0 [27.7-30.5] weeks; 52 boys [55%]; median [IQR] birth weight, 1250 [1020-1445] g; placebo group: median [IQR] GA, 28.8 [27.6-30.4] weeks; 54 boys [55%]; median [IQR] birth weight, 1208 [1021-1430] g). The data safety monitoring board advised to discontinue the study early based on interim futility analysis (including the first 225 randomized infants), as the conditional power did not reach the prespecified threshold of 35% for both rh-insulin dosages. The study continued while the data safety monitoring board analyzed and discussed the data. In the final intention-to-treat analysis, the median (IQR) time to achieve FEF was significantly reduced in 94 infants receiving low-dose rh insulin (10.0 [7.0-21.8] days; P = .03) and in 82 infants receiving high-dose rh insulin (10.0 [6.0-15.0] days; P = .001) compared with 85 infants receiving placebo (14.0 [8.0-28.0] days). Compared with placebo, the difference in median (95% CI) time to FEF was 4.0 (1.0-8.0) days for the low-dose group and 4.0 (1.0-7.0) days for the high-dose group. Weight gain rates did not differ significantly between groups. Necrotizing enterocolitis (Bell stage 2 or 3) occurred in 7 of 108 infants (6%) in the low-dose group, 4 of 88 infants (5%) in the high-dose group, and 10 of 97 infants (10%) in the placebo group. None of the infants developed serum insulin antibodies. CONCLUSIONS AND RELEVANCE Results of this randomized clinical trial revealed that enteral administration of 2 different rh-insulin dosages was safe and compared with placebo, significantly reduced time to FEF in preterm infants with a GA of 26 to 32 weeks. These findings support the use of rh insulin as a supplement to human milk and preterm formula. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02510560.
Collapse
Affiliation(s)
- Elise Mank
- Department of Pediatrics-Neonatology, Amsterdam UMC, University of Amsterdam, Vrije Universiteit Amsterdam, Emma Children’s Hospital, Amsterdam, the Netherlands
| | - Miguel Sáenz de Pipaón
- Department of Pediatrics-Neonatology, La Paz University Hospital, Autonoma University of Madrid, Madrid, Spain
| | - Alexandre Lapillonne
- Department of Neonatology, Assistance Publique–Hôpitaux de Paris Necker-Enfants Malades Hospital, Paris University EHU 7328, Paris, France
| | - Virgilio P. Carnielli
- Department of Pediatrics-Neonatology, Ospedali Riuniti di Ancona, Polytechnic University of Marche, Azienda Ospedaliero Universitaria, Ancona, Italy
| | - Thibault Senterre
- Department of Pediatrics-Neonatology, Centre Hospitalier Régional de la Citadelle, University of Liège, Liège, Belgium
| | - Raanan Shamir
- Schneider Children’s Medical Center of Israel, Petah Tikva, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Letty van Toledo
- Department of Pediatrics-Neonatology, Amsterdam UMC, University of Amsterdam, Vrije Universiteit Amsterdam, Emma Children’s Hospital, Amsterdam, the Netherlands
| | - Johannes B. van Goudoever
- Department of Pediatrics-Neonatology, Amsterdam UMC, University of Amsterdam, Vrije Universiteit Amsterdam, Emma Children’s Hospital, Amsterdam, the Netherlands
| | | |
Collapse
|
5
|
How to Provide Breast Milk for the Preterm Infant and Avoid Symptomatic Cytomegalovirus Infection with Possible Long-Term Sequelae. Life (Basel) 2022; 12:life12040504. [PMID: 35454995 PMCID: PMC9031638 DOI: 10.3390/life12040504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/04/2022] Open
Abstract
Cytomegalovirus (CMV) is able to replicate in the breast milk of lactating mothers and thus the offspring might be affected by mild to severe symptoms of postnatal CMV disease in case of prematurity; not in term infants. Sepsis-like syndrome affects only very low birth infants; and few cases have been reported. The neurodevelopmental long-term outcome of those preterm infants revealed possible subtle deficiencies, but no major neurodevelopmental impairment. Neurodevelopmental sequelae are still in discussion and seem somewhat overestimated after careful evaluation of the published evidence. The main focus of postnatal CMV disease lies upon the extremely low birth weight of infants. Elimination of CMV is provided by short-term heating methods like the most widely used Holder pasteurization. Freezing and thawing methods leave a risk for CMV acquisition. The benefits of untreated breast milk have to be considered to outweigh the possible sequelae of postnatal CMV infection in the most vulnerable preterm infants.
Collapse
|
6
|
Development of the Gastrointestinal Tract in Newborns as a Challenge for an Appropriate Nutrition: A Narrative Review. Nutrients 2022; 14:nu14071405. [PMID: 35406018 PMCID: PMC9002905 DOI: 10.3390/nu14071405] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023] Open
Abstract
The second and third trimesters of pregnancy are crucial for the anatomical and functional development of the gastrointestinal (GI) tract. If premature birth occurs, the immaturity of the digestive and absorptive processes and of GI motility represent a critical challenge to meet adequate nutritional needs, leading to poor extrauterine growth and to other critical complications. Knowledge of the main developmental stages of the processes involved in the digestion and absorption of proteins, carbohydrates, and lipids, as well as of the maturational phases underlying the development of GI motility, may aid clinicians to optimize the nutritional management of preterm infants. The immaturity of these GI systems and functions may negatively influence the patterns of gut colonization, predisposing to an abnormal microbiome. This, in turn, further contributes to alter the functional, immune, and neural development of the GI tract and, especially in preterm infants, has been associated with an increased risk of severe GI complications, such as necrotizing enterocolitis. Deeper understanding of the physiological colonization patterns in term and preterm infants may support the promotion of these patterns and the avoidance of microbial perturbations associated with the development of several diseases throughout life. This review aims to provide a global overview on the maturational features of the main GI functions and on their implications following preterm birth. We will particularly focus on the developmental differences in intestinal digestion and absorption functionality, motility, gut–brain axis interaction, and microbiomes.
Collapse
|
7
|
Mank E, van Toledo L, Heijboer AC, van den Akker CHP, van Goudoever JB. Insulin Concentration in Human Milk in the First Ten Days Postpartum: Course and Associated Factors. J Pediatr Gastroenterol Nutr 2021; 73:e115-e119. [PMID: 34183615 DOI: 10.1097/mpg.0000000000003214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND OBJECTIVES Human milk (HM) is better tolerated than formula in preterm infants. Insulin, which is naturally present in HM but not in formula, has been suggested as a key factor for feeding tolerance, as it appears to stimulate intestinal maturation. Its precise concentrations during the early postnatal period, however, remains unknown. The objective of this study was to assess the natural timecourse of the HM insulin concentration during the first ten days postpartum. The effect of preterm delivery, maternal obesity, and diurnal rhythm were also assessed. METHOD HM was collected from 31 non-diabetic mothers (21 preterm [gestational age (GA) < 37 weeks]; 10 at-term [GA ≥ 37 weeks]) on ≥ 4 time-points per day during the first five days, and once on the tenth day postpartum. RESULTS The HM insulin concentration declined rapidly within the first three days postpartum (day 1: 516 [312-1058] pmol/L; day 3: 157 [87-299] pmol/L), after which the concentration remained relatively stable. The insulin concentrations were higher in HM from obese mothers than from non-obese mothers (P < 0.001). Preterm delivery did not significantly affect HM insulin concentrations when adjusted for maternal pre-pregnancy body mass index category (P = 0.270). Diurnal rhythm was characterized by an insulin concentration decline throughout the night (P = 0.001), followed by an increase in the morning (P = 0.001). CONCLUSION The HM insulin concentration declines rapidly in the first three days postpartum, follows a diurnal rhythm, and is higher in obese mothers compared to non-obese mothers. HM insulin concentrations are not affected by preterm delivery.
Collapse
Affiliation(s)
- Elise Mank
- Department of Pediatrics-Neonatology, Emma Children's Hospital
| | | | - Annemieke C Heijboer
- Department of Clinical Chemistry, Endocrine Laboratory, Amsterdam Gastroenterology Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
8
|
Abramov VM, Kosarev IV, Priputnevich TV, Machulin AV, Abashina TN, Chikileva IO, Donetskova AD, Takada K, Melnikov VG, Vasilenko RN, Khlebnikov VS, Samoilenko VA, Nikonov IN, Sukhikh GT, Uversky VN, Karlyshev AV. S-layer protein 2 of vaginal Lactobacillus crispatus 2029 enhances growth, differentiation, VEGF production and barrier functions in intestinal epithelial cell line Caco-2. Int J Biol Macromol 2021; 189:410-419. [PMID: 34437917 DOI: 10.1016/j.ijbiomac.2021.08.150] [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: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
We have previously demonstrated the ability of the human vaginal strain Lactobacillus crispatus 2029 (LC2029) for strong adhesion to cervicovaginal epithelial cells, expression of the surface layer protein 2 (Slp2), and antagonistic activity against urogenital pathogens. Slp2 forms regular two-dimensional structure around the LC2029 cells,which is secreted into the medium and inhibits intestinal pathogen-induced activation of caspase-9 and caspase-3 in the human intestinal Caco-2 cells. Here, we elucidated the effects of soluble Slp2 on adhesion of proteobacteria pathogens inducing necrotizing enterocolitis (NEC), such as Escherichia coli ATCC E 2348/69, E. coli ATCC 31705, Salmonella Enteritidis ATCC 13076, Campylobacter jejuni ATCC 29428, and Pseudomonas aeruginosa ATCC 27853 to Caco-2 cells, as well as on growth promotion, differentiation, vascular endothelial growth factor (VEGF) production, and intestinal barrier function of Caco-2 cell monolayers. Slp2 acts as anti-adhesion agent for NEC-inducing proteobacteria, promotes growth of immature Caco-2 cells and their differentiation, and enhances expression and functional activity of sucrase, lactase, and alkaline phosphatase. Slp2 stimulates VEGF production, decreases paracellular permeability, and increases transepithelial electrical resistance, strengthening barrier function of Caco-2 cell monolayers. These data support the important role of Slp2 in the early postnatal development of the human small intestine enterocytes.
Collapse
Affiliation(s)
- Vyacheslav M Abramov
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Igor V Kosarev
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Tatiana V Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Andrey V Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Tatiana N Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Irina O Chikileva
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Laboratory of Cell Immunity, Blokhin National Research, Center of Oncology Ministry of Health RF, Moscow 115478, Russia
| | | | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Vyacheslav G Melnikov
- Gabrichevsky Moscow Research Institute of Epidemiology and Microbiology, Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow 152212, Russia
| | - Raisa N Vasilenko
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia
| | | | - Vladimir A Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Ilya N Nikonov
- Federal State Education Institution of Higher Professional Education Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, Moscow 109472, Russia
| | - Gennady T Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Andrey V Karlyshev
- Department of Science, Engineering and Computing, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
9
|
Picaud JC, De Magistris A, Mussap M, Corbu S, Dessì A, Noto A, Fanos V, Cesare Marincola F. Urine NMR Metabolomics Profile of Preterm Infants With Necrotizing Enterocolitis Over the First Two Months of Life: A Pilot Longitudinal Case-Control Study. Front Mol Biosci 2021; 8:680159. [PMID: 34212004 PMCID: PMC8239193 DOI: 10.3389/fmolb.2021.680159] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/20/2021] [Indexed: 12/22/2022] Open
Abstract
Objective: To investigate changes in the urine metabolome of very low birth weight preterm newborns with necrotizing enterocolitis (NEC) and feed intolerance, we conducted a longitudinal study over the first 2 months of life. The metabolome of NEC newborns was compared with two control groups that did not develop NEC: the first one included preterm babies with feed intolerance, while the second one preterm babies with good feed tolerance. Methods: Newborns developing NEC within the 3 weeks of life were identified as early onset NEC, while the remaining as late onset NEC. Case-control matching was done according to the gestational age (±1 week), birth weight (± 200 g), and postnatal age. A total of 96 urine samples were collected and analyzed. In newborns with NEC, samples were collected before, during and after the diagnosis over the first 2 months of life, while in controls samples were collected as close as possible to the postnatal age of newborns with NEC. Proton nuclear magnetic resonance (1H NMR) spectroscopy was used for metabolomic analysis. Data were analyzed by univariate and multivariate statistical analysis. Results: In all the preterm newborns, urine levels of betaine, glycine, succinate, and citrate positively correlated with postnatal age. Suberate and lactate correlated with postnatal age in preterms with NEC and in controls with food intolerance, while N,N-dimethylglycine (N,N-DMG) correlated only in controls with good digestive tolerance. Preterm controls with feed intolerance showed a progressive significant decrease of N-methylnicotinamide and carnitine. Lactate, betaine, myo-inositol, urea, creatinine, and N,N-dimethylglycine discriminated late-onset NEC from controls with good feed tolerance. Conclusion: Our findings are discussed in terms of contributions from nutritional and clinical managements of patients and gut microbiota.
Collapse
Affiliation(s)
- Jean-Charles Picaud
- Neonatology Unit, Croix-Rousse University Hospital, Hospices Civils de Lyon, Lyon, France
| | - Anna De Magistris
- Pediatrics and Neonatology Division of, Azienda USL Romagna, Santa Maria Delle Croci Hospital, Ravenna, Italy
| | - Michele Mussap
- Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Sara Corbu
- Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Angelica Dessì
- Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonio Noto
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Vassilios Fanos
- Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Flaminia Cesare Marincola
- Department of Chemical and Geological Sciences, Cittadella Universitaria di Monserrato, University of Cagliari, Cagliari, Italy
| |
Collapse
|
10
|
Mank E, van Harskamp D, van Toledo L, van Goudoever JB, Schierbeek H. Simultaneous assessment of intestinal permeability and lactase activity in human-milk-fed preterm infants by sugar absorption test: Clinical implementation and analytical method. Clin Nutr 2020; 40:1413-1419. [PMID: 32948350 DOI: 10.1016/j.clnu.2020.08.034] [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: 04/26/2020] [Revised: 05/25/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND & AIMS Experimental (nutritional) interventions in preterm infants frequently focus on intestinal maturation, as improving tolerance to enteral nutrition is a major goal. Intestinal permeability and lactase activity serve as markers for intestinal maturation. We aimed to develop a protocol for the simultaneous assessment of both markers in human-milk-fed preterm infants by a sugar absorption test. In addition, we developed a new gas chromatography-mass spectrometry (GC-MS) method for the analysis of lactulose, lactose, and mannitol in urine and milk collected during the sugar absorption test. METHODS The sugar absorption test was performed on days 4, 7, and 14 postpartum in 12 preterm infants (gestational age of 26-32 weeks). Human milk was collected, pooled, and divided into equal portions to provide a stable lactose intake for 24 h. Urine was collected in the last 6 h of this 24 h period, after administration of a bolus test sugar solution. Samples were analyzed by GC-MS after derivatization by oxime formation combined with acetylation. RESULTS The GC-MS method was validated and used for the accurate measurement of lactulose, lactose, and mannitol concentrations. The urinary lactulose/mannitol ratio declined with time, suggesting a decreased intestinal permeability. The urine-to-milk-lactulose/lactose ratio increased as a result of increased lactase activity with time. CONCLUSIONS The developed protocol for simultaneous assessment of intestinal permeability and lactase activity can be used to monitor the effect of experimental (nutritional) interventions in human-milk-fed preterm infants. Urine and milk samples obtained during the sugar absorption test can be accurately analyzed by GC-MS.
Collapse
Affiliation(s)
- Elise Mank
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Dewi van Harskamp
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Stable Isotope Research Laboratory, Endocrinology, Amsterdam Gastroenterology Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Letty van Toledo
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Johannes B van Goudoever
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| | - Henk Schierbeek
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Stable Isotope Research Laboratory, Endocrinology, Amsterdam Gastroenterology Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Abstract
The human fetus receives oral nutrition through swallowed amniotic fluid and this makes a significant nutritional contribution to the fetus. Postnatally, macronutrient absorption and digestion appear to function well in the preterm infant. Although pancreatic function is relatively poor, the newborn infant has several mechanisms to overcome this. These include a range of digestive enzymes in human milk, novel digestive enzymes involved in fat and protein digestion that do not appear to be present in the older child or adult, and the presence of a Bifidobacterium-rich colonic microbiome that may "scavenge" unabsorbed macronutrients and make them available to the infant.
Collapse
Affiliation(s)
- Marta Rogido
- Goryeb Children's Hospital, Morristown, NJ.,Mid-Atlantic Neonatal Associates, Morristown, NJ.,Biomedical Research Institute of New Jersey, Cedar Knolls, NJ
| | - Ian Griffin
- Goryeb Children's Hospital, Morristown, NJ.,Mid-Atlantic Neonatal Associates, Morristown, NJ.,Biomedical Research Institute of New Jersey, Cedar Knolls, NJ
| |
Collapse
|
12
|
Jena A, Montoya CA, Mullaney JA, Dilger RN, Young W, McNabb WC, Roy NC. Gut-Brain Axis in the Early Postnatal Years of Life: A Developmental Perspective. Front Integr Neurosci 2020; 14:44. [PMID: 32848651 PMCID: PMC7419604 DOI: 10.3389/fnint.2020.00044] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that alterations in the development of the gastrointestinal (GI) tract during the early postnatal period can influence brain development and vice-versa. It is increasingly recognized that communication between the GI tract and brain is mainly driven by neural, endocrine, immune, and metabolic mediators, collectively called the gut-brain axis (GBA). Changes in the GBA mediators occur in response to the developmental changes in the body during this period. This review provides an overview of major developmental events in the GI tract and brain in the early postnatal period and their parallel developmental trajectories under physiological conditions. Current knowledge of GBA mediators in context to brain function and behavioral outcomes and their synthesis and metabolism (site, timing, etc.) is discussed. This review also presents hypotheses on the role of the GBA mediators in response to the parallel development of the GI tract and brain in infants.
Collapse
Affiliation(s)
- Ankita Jena
- School of Food & Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand.,The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Carlos A Montoya
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Jane A Mullaney
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Wayne Young
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Warren C McNabb
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
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: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
14
|
Romero-Velarde E, Delgado-Franco D, García-Gutiérrez M, Gurrola-Díaz C, Larrosa-Haro A, Montijo-Barrios E, Muskiet FAJ, Vargas-Guerrero B, Geurts J. The Importance of Lactose in the Human Diet: Outcomes of a Mexican Consensus Meeting. Nutrients 2019; 11:E2737. [PMID: 31718111 PMCID: PMC6893676 DOI: 10.3390/nu11112737] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022] Open
Abstract
Lactose is a unique component of breast milk, many infant formulas and dairy products, and is widely used in pharmaceutical products. In spite of that, its role in human nutrition or lactose intolerance is generally not well-understood. For that reason, a 2-day-long lactose consensus meeting with health care professionals was organized in Mexico to come to a set of statements for which consensus could be gathered. Topics ranging from lactase expression to potential health benefits of lactose were introduced by experts, and that was followed by a discussion on concept statements. Interestingly, lactose does not seem to induce a neurological reward response when consumed. Although lactose digestion is optimal, it supplies galactose for liver glycogen synthesis. In infants, it cannot be ignored that lactose-derived galactose is needed for the synthesis of glycosylated macromolecules. At least beyond infancy, the low glycemic index of lactose might be metabolically beneficial. When lactase expression decreases, lactose maldigestion may lead to lactose intolerance symptoms. In infancy, the temporary replacing of lactose by other carbohydrates is only justified in case of severe intolerance symptoms. In those who show an (epi)genetic decrease or absence of lactase expression, a certain amount (for adults mostly up to 12 g per portion) of lactose can still be consumed. In these cases, lactose shows beneficial intestinal-microbiota-shaping effects. Avoiding lactose-containing products may imply a lower intake of other important nutrients, such as calcium and vitamin B12 from dairy products, as well as an increased intake of less beneficial carbohydrates.
Collapse
Affiliation(s)
- Enrique Romero-Velarde
- Instituto de Nutrición Humana, Universidad de Guadalajara and Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, 44340 Guadalajara, Jalisco, Mexico
| | - Dagoberto Delgado-Franco
- Neonatology Department. ABC Medical Center, 01120 Mexico City and Instituto Tecnológico de Estudios Superiores de Monterrey, 64849 Monterrey, Mexico;
| | | | - Carmen Gurrola-Díaz
- Departamento de Biología Molecular y Genómica. Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, 44340 Guadalajara, Jalisco, Mexico; (C.G.-D.); (B.V.-G.)
| | - Alfredo Larrosa-Haro
- Instituto de Nutrición Humana, Universidad de Guadalajara, 44340 Guadalajara, Jalisco, Mexico;
| | - Ericka Montijo-Barrios
- Servicio de Gastroenterología. Instituto Nacional de Pediatría, 04530 Mexico City, Mexico;
| | - Frits A. J. Muskiet
- Laboratory Medicine, University Medical Center Groningen and University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Belinda Vargas-Guerrero
- Departamento de Biología Molecular y Genómica. Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, 44340 Guadalajara, Jalisco, Mexico; (C.G.-D.); (B.V.-G.)
| | - Jan Geurts
- FrieslandCampina, 3818 LEAmersfoort, The Netherlands;
| |
Collapse
|
15
|
Knopp JL, Signal M, Harris DL, Marics G, Weston P, Harding J, Tóth-Heyn P, Hómlok J, Benyó B, Chase JG. Modelling intestinal glucose absorption in premature infants using continuous glucose monitoring data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2019; 171:41-51. [PMID: 30344050 DOI: 10.1016/j.cmpb.2018.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 09/11/2018] [Accepted: 10/01/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Model-based glycaemic control protocols have shown promise in neonatal intensive care units (NICUs) for reducing both hyperglycaemia and insulin-therapy driven hypoglycaemia. However, current models for the appearance of glucose from enteral feeding are based on values from adult intensive care cohorts. This study aims to determine enteral glucose appearance model parameters more reflective of premature infant physiology. METHODS Peaks in CGM data associated with enteral milk feeds in preterm and term infants are used to fit a two compartment gut model. The first compartment describes glucose in the stomach, and the half life of gastric emptying is estimated as 20 min from literature. The second compartment describes glucose in the small intestine, and absorption of glucose into the blood is fit to CGM data. Two infant cohorts from two NICUs are used, and results are compared to appearances derived from data in highly controlled studies in literature. RESULTS The average half life across all infants for glucose absorption from the gut to the blood was 50 min. This result was slightly slower than, but of similar magnitude to, results derived from literature. No trends were found with gestational or postnatal age. Breast milk fed infants were found to have a higher absorption constant than formula fed infants, a result which may reflect known differences in gastric emptying for different feed types. CONCLUSIONS This paper presents a methodology for estimation of glucose appearance due to enteral feeding, and model parameters suitable for a NICU model-based glycaemic control context.
Collapse
Affiliation(s)
- J L Knopp
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand.
| | - M Signal
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand.
| | - D L Harris
- Newborn Intensive Care Unit, Waikato District Health Board, Hamilton, New Zealand; Liggins Institute, University of Auckland, Auckland, New Zealand.
| | - G Marics
- First Department of Paediatrics, Intensive Care Unit, Semmelweis University, Budapest, Hungary
| | - P Weston
- Newborn Intensive Care Unit, Waikato District Health Board, Hamilton, New Zealand.
| | - J Harding
- Liggins Institute, University of Auckland, Auckland, New Zealand.
| | - P Tóth-Heyn
- First Department of Paediatrics, Intensive Care Unit, Semmelweis University, Budapest, Hungary.
| | - J Hómlok
- Budapest University of Technology and Economics, Budapest, Hungary
| | - B Benyó
- Budapest University of Technology and Economics, Budapest, Hungary.
| | - J G Chase
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand.
| |
Collapse
|
16
|
Neal-Kluever A, Fisher J, Grylack L, Kakiuchi-Kiyota S, Halpern W. Physiology of the Neonatal Gastrointestinal System Relevant to the Disposition of Orally Administered Medications. Drug Metab Dispos 2018; 47:296-313. [DOI: 10.1124/dmd.118.084418] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
|
17
|
Lenfestey MW, Neu J. Gastrointestinal Development: Implications for Management of Preterm and Term Infants. Gastroenterol Clin North Am 2018; 47:773-791. [PMID: 30337032 DOI: 10.1016/j.gtc.2018.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) system provides digestive, absorptive, neuroendocrine, and immunologic functions to support overall health. If normal development is interrupted, a variety of complications and disease can arise. This article explores normal development of the GI tract and specific clinical challenges pertinent to preterm and term infants. Specific topics include abnormal motility, gastroesophageal reflux, current feeding recommendations for preterm infants, effects of parenteral nutrition, and the relationship between the GI tract and the immune system.
Collapse
Affiliation(s)
- Mary W Lenfestey
- Department of Pediatrics, University of Florida, PO Box 100296, Gainesville, FL 32610, USA
| | - Josef Neu
- Department of Pediatrics, University of Florida, 6516 Southwest 93rd Avenue, Gainesville, FL 32610, USA.
| |
Collapse
|
18
|
Sun J, Li Y, Pan X, Nguyen DN, Brunse A, Bojesen AM, Rudloff S, Mortensen MS, Burrin DG, Sangild PT. Human Milk Fortification with Bovine Colostrum Is Superior to Formula-Based Fortifiers to Prevent Gut Dysfunction, Necrotizing Enterocolitis, and Systemic Infection in Preterm Pigs. JPEN J Parenter Enteral Nutr 2018; 43:252-262. [PMID: 29992630 DOI: 10.1002/jpen.1422] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Fortification of donor human milk (DHM) is required for optimal growth of very preterm infants, but there are concerns of more gut dysfunction and necrotizing enterocolitis (NEC) when using formula-based fortifiers (FFs), especially soon after birth. Intact bovine colostrum (BC) is rich in nutrients and bioactive factors, and protects against NEC in preterm pigs. We hypothesized that fortification of DHM with BC is superior to FFs to prevent gut dysfunction and infections when provided shortly after preterm birth. METHODS Two FF products, Enfamil (ENF; intact protein, vegetable oil) and PreNAN+Nutrilon (NAN; extensively hydrolyzed protein, maltodextrin), were compared with BC as fortifier to DHM fed to preterm pigs for 5 days. RESULTS Relative to the DHM+BC group, DHM+FF groups had higher diarrhea score and lower hexose uptake and lactase activity, and specifically the DHM+NAN group showed higher gut permeability, NEC score, more mucosa-adherent bacteria with altered gut microbiota structure (ie, lower diversity, increased Enterococcus, decreased Staphylococcus abundance). Both DHM+FF groups showed higher expression of intestinal cytokine and inflammation-related genes, more gut-derived bacteria in the bone marrow, lower density of mucin-containing goblet cells, and slightly higher colon lactate, stomach pH and acetate, and blood neutrophil-to-lymphocyte levels than the DHM+BC group. CONCLUSIONS Used as a fortifier to DHM, BC is superior to FFs to support gut function, nutrient absorption, and bacterial defense mechanisms in preterm pigs. It is important to optimize the composition of nutrient fortifiers for preterm infants fed human milk.
Collapse
Affiliation(s)
- Jing Sun
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yanqi Li
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaoyu Pan
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Brunse
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders M Bojesen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Silvia Rudloff
- Institute of Nutritional Science, Justus-Liebig-University Giessen , Giessen, Germany
| | | | - Douglas G Burrin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Per T Sangild
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
19
|
Call L, Stoll B, Oosterloo B, Ajami N, Sheikh F, Wittke A, Waworuntu R, Berg B, Petrosino J, Olutoye O, Burrin D. Metabolomic signatures distinguish the impact of formula carbohydrates on disease outcome in a preterm piglet model of NEC. MICROBIOME 2018; 6:111. [PMID: 29921329 PMCID: PMC6009052 DOI: 10.1186/s40168-018-0498-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/08/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Major risk factors for necrotizing enterocolitis (NEC) include premature birth and formula feeding in the context of microbial colonization of the gastrointestinal tract. We previously showed that feeding formula composed of lactose vs. corn syrup solids protects against NEC in preterm pigs; however, the microbial and metabolic effects of these different carbohydrates used in infant formula has not been explored. OBJECTIVE Our objective was to characterize the effects of lactose- and corn syrup solid-based formulas on the metabolic and microbial profiles of preterm piglets and to determine whether unique metabolomic or microbiome signatures correlate with severity or incidence of NEC. DESIGN/METHODS Preterm piglets (103 days gestation) were given total parenteral nutrition (2 days) followed by gradual (5 days) advancement of enteral feeding of formulas matched in nutrient content but containing either lactose (LAC), corn syrup solids (CSS), or 1:1 mix (MIX). Gut contents and mucosal samples were collected and analyzed for microbial profiles by sequencing the V4 region of the 16S rRNA gene. Metabolomic profiles of cecal contents and plasma were analyzed by LC/GC mass spectrometry. RESULTS NEC incidence was 14, 50, and 44% in the LAC, MIX, and CSS groups, respectively. The dominant classes of bacteria were Bacilli, Clostridia, and Gammaproteobacteria. The number of observed OTUs was lowest in colon contents of CSS-fed pigs. CSS-based formula was associated with higher Bacilli and lower Clostridium from clusters XIVa and XI in the colon. NEC was associated with decreased Gammaproteobacteria in the stomach and increased Clostridium sensu stricto in the ileum. Plasma from NEC piglets was enriched with metabolites of purine metabolism, aromatic amino acid metabolism, and bile acids. Markers of glycolysis, e.g., lactate, were increased in the cecal contents of CSS-fed pigs and in plasma of pigs which developed NEC. CONCLUSIONS Feeding formula containing lactose is not completely protective against NEC, yet selects for greater microbial richness associated with changes in Bacilli and Clostridium and lower NEC incidence. We conclude that feeding preterm piglets a corn syrup solid vs. lactose-based formula increases the incidence of NEC and produces distinct metabolomic signatures despite modest changes in microbiome profiles.
Collapse
Affiliation(s)
- Lee Call
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| | - Barbara Stoll
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| | - Berthe Oosterloo
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| | - Nadim Ajami
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, One Baylor Plaza, MS BCM385, Houston, TX 77030 USA
| | - Fariha Sheikh
- Division of Pediatric Surgery, Baylor College of Medicine, 6701 Fannin St, Suite 1210, Houston, TX 77030 USA
| | - Anja Wittke
- Mead Johnson Pediatric Nutrition Institute, 2400 W Lloyd Expressway, Evansville, IN 47712 USA
| | - Rosaline Waworuntu
- Mead Johnson Pediatric Nutrition Institute, 2400 W Lloyd Expressway, Evansville, IN 47712 USA
| | - Brian Berg
- Mead Johnson Pediatric Nutrition Institute, 2400 W Lloyd Expressway, Evansville, IN 47712 USA
| | - Joseph Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, One Baylor Plaza, MS BCM385, Houston, TX 77030 USA
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Baylor College of Medicine, 6701 Fannin St, Suite 1210, Houston, TX 77030 USA
| | - Douglas Burrin
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| |
Collapse
|
20
|
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
|
21
|
Ontsouka EC, Albrecht C, Bruckmaier RM. Invited review: Growth-promoting effects of colostrum in calves based on interaction with intestinal cell surface receptors and receptor-like transporters. J Dairy Sci 2016; 99:4111-4123. [PMID: 26874414 DOI: 10.3168/jds.2015-9741] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/28/2015] [Indexed: 12/13/2022]
Abstract
The postnatal development and maturation of the gastrointestinal (GI) tract of neonatal calves is crucial for their survival. Major morphological and functional changes in the calf's GI tract initiated by colostrum bioactive substances promote the establishment of intestinal digestion and absorption of food. It is generally accepted that colostrum intake provokes the maturation of organs and systems in young calves, illustrating the significance of the cow-to-calf connection at birth. These postnatal adaptive changes of the GI tissues in neonatal calves are especially induced by the action of bioactive substances such as insulin-like growth factors, hormones, or cholesterol carriers abundantly present in colostrum. These substances interact with specific cell-surface receptors or receptor-like transporters expressed in the GI wall of neonatal calves to elicit their biological effects. Therefore, the abundance and activity of cell surface receptors and receptor-like transporters binding colostral bioactive substances are a key aspect determining the effects of the cow-to-calf connection at birth. The present review compiles the information describing the effects of colostrum feeding on selected serum metabolic and endocrine traits in neonatal calves. In this context, the current paper discusses specifically the consequences of colostrum feeding on the GI expression and activity of cell-receptors and receptor-like transporters binding growth hormone, insulin-like growth factors, insulin, or cholesterol acceptors in neonatal calves.
Collapse
Affiliation(s)
- Edgar C Ontsouka
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, CH-3012 Bern, Switzerland; Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland.
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, CH-3012 Bern, Switzerland; Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Rupert M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| |
Collapse
|
22
|
|
23
|
Lai S, Yu W, Wallace L, Sigalet D. Intestinal muscularis propria increases in thickness with corrected gestational age and is focally attenuated in patients with isolated intestinal perforations. J Pediatr Surg 2014; 49:114-9. [PMID: 24439593 DOI: 10.1016/j.jpedsurg.2013.09.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 09/30/2013] [Indexed: 11/16/2022]
Abstract
PURPOSE Intestinal perforations are common in premature infants, leading to a diagnostic dilemma between necrotizing enterocolitis and isolated intestinal perforation (IIP). IIP is thought to result from a congenital or acquired absence of the muscularis propria. However, developmental events leading to IIP are not well understood. This study examines the relationship between corrected gestational age (CGA) and intestinal muscle development in controls and patients with IIP. METHODS Specimens from stillbirths and infants undergoing intestinal surgery from 8 to 48weeks' CGA were collected from 2005 to 2012. Twelve patients with IIP were identified. Control specimens were collected during 25 fetal autopsies and 39 bowel resections. In each case, three sections of intestine were examined histologically for muscularis mucosa, circular and longitudinal muscle thickness. Comparisons of control and perforated specimens were performed via linear regression and ANOVA. RESULTS Controls and adjacent normal segments in IIP showed a linear relationship between thickness of circular and longitudinal muscles with CGA. Circular and longitudinal muscles were thinner in perforated segments than in adjacent normals and CGA-matched controls (p<0.05). CONCLUSION Intestinal muscularis propria increases in thickness with CGA. Muscle thickness is focally attenuated in patients with isolated intestinal perforations, while the remaining intestine is normal, suggesting that primary repair is an appropriate treatment.
Collapse
Affiliation(s)
- Sarah Lai
- Division of Pediatic Surgery, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Weiming Yu
- Department of Pathology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Laurie Wallace
- Division of Pediatic Surgery, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - David Sigalet
- Division of Pediatic Surgery, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
24
|
An innovative investigation into the etiology of feeding intolerance in preterm infants. J Pediatr Gastroenterol Nutr 2014; 58:4-5. [PMID: 24121141 DOI: 10.1097/mpg.0000000000000188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Li Y, Jensen ML, Chatterton DEW, Jensen BB, Thymann T, Kvistgaard AS, Sangild PT. Raw bovine milk improves gut responses to feeding relative to infant formula in preterm piglets. Am J Physiol Gastrointest Liver Physiol 2014; 306:G81-90. [PMID: 24157971 DOI: 10.1152/ajpgi.00255.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
For preterm neonates, the quality of the first milk is crucial for intestinal maturation and resistance to necrotizing enterocolitis (NEC). Among other factors, milk quality is determined by the stage of lactation and processing. We hypothesized that unprocessed mature bovine milk (BM; raw bovine milk) would have less bioactivity than corresponding bovine colostrum (BC) in a preterm pig model, but have improved bioactivity relative to its homogenized, pasteurized, spray-dried equivalent, whole milk powder (WMP), or a bovine milk protein-based infant formula (IF). For 5 days, newborn preterm pigs received parenteral and enteral nutrition consisting of IF (n = 13), BM (n = 13), or BC (n = 14). In a second study, WMP (n = 15) was compared with IF (n = 10) and BM (n = 9). Compared with pigs fed IF, pigs that were fed BM had significantly improved intestinal structure (mucosal weight, villus height) and function (increased nutrient absorption and enzyme activities, decreased gut permeability, nutrient fermentation, and NEC severity). BC further improved these effects relative to BM (lactase activity, lactose absorption, plasma citrulline, and tissue interleukin-8). WMP induced similar effects as BM, except for lactase activity and lactose absorption. In conclusion, the maturational and protective effects on the immature intestine decreased in the order BC>BM>WMP, but all three intact bovine milk diets were markedly better than IF. The stage of lactation (colostrum vs. mature milk) and milk processing (e.g., homogenization, fractionation, pasteurization, spray-drying) are important factors in determining milk quality during the early postnatal period of preterm neonates.
Collapse
Affiliation(s)
- Yanqi Li
- 30 Rolighedsvej, DK-1958 Frederiksberg C, Denmark.
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Feeding intolerance (FI), defined as the inability to digest enteral feedings associated to increased gastric residuals, abdominal distension and/or emesis, is frequently encountered in the very preterm infant and often leads to a disruption of the feeding plan. In most cases FI represents a benign condition related to the immaturity of gastrointestinal function, however its presentation may largely overlap with that of an impending necrotizing enterocolitis. As a consequence, individual interpretation of signs of FI represents one of the most uncontrollable variables in the early nutritional management of these infants, and may lead to suboptimal nutrition, delayed attainment of full enteral feeding and prolonged intravenous nutrition supply. Strategies aimed at preventing and/or treating FI are diverse, although very few have been validated in large RCT and systematic reviews. The purpose of this paper is to summarize the existing information on this topic, spanning from patho-physiological and clinical aspects to the prevention and treatment strategies tested in clinical studies, with specific attention to practical issues.
Collapse
|
27
|
Abstract
OBJECTIVES Feeding intolerance (FI) in preterm infants is common but the etiology remains unclear. This study examined FI as a stress-related disease involving brain-gut interactions and tested the model of allostatic load and complications of prematurity. Specific aims were to describe demographic/medical variables and biomarker levels at each time and over time for the sample; describe/compare variables and biomarker levels at each time for infants with/without FI; and compare biomarker interquartile/interpercentile distributions between infants with/without FI. METHODS Preterm infants <32 weeks' gestation were recruited. The primary outcome was FI by day 7 defined as a feeding withheld, discontinued, or decreased because the infant was not tolerating enteral feedings. Allostatic load was operationalized using cortisol and 8-hydroxydeoxyguanosine (8-OHdG) from cord blood and from saliva and urine on days 1, 7, and 14. Descriptive statistics and comparative analyses were performed. RESULTS Seven of 31 infants enrolled met criteria for FI. Infants with FI had lower median urinary cortisol on day 1 (P = 0.007) and trended to have lower cortisol in the cord blood (P = 0.056). Interquartile distributions were significantly different between infants with/without FI for urinary cortisol on day 1 (P = 0.034) and trended for differences in 8-OHdG on day 14 (P = 0.087). Interpercentile distributions were significantly different in salivary cortisol on day 14 (P = 0.034) and trended for differences in 8-OHdG on day 1 (P = 0.079). CONCLUSIONS Results support further testing of the model in a larger sample; investigation of the cellular mechanisms associated with the stress and the free radical/antioxidant systems; and inclusion of prenatal factors.
Collapse
|
28
|
Kohler JA, Perkins AM, Bass WT. Human milk versus formula after gastroschisis repair: effects on time to full feeds and time to discharge. J Perinatol 2013; 33:627-30. [PMID: 23519369 DOI: 10.1038/jp.2013.27] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 02/15/2013] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To determine if the choice of enteral feeds after gastroschisis repair relates to the time to achieve full feeds and time to discharge. STUDY DESIGN A retrospective study of infants with gastroschisis from 2000 to 2010 examined demographics, days at closure, days at initiation of feeds, days to full feeds, time to discharge and length of stay. RESULT Ninety infants were identified, 22 received (human milk) HM exclusively, 15 were fed >50% HM, 16 were fed <50% HM and 26 were fed only cow milk-based formulas. Infants fed exclusively HM had significantly shorter times to full enteral feedings (median 5 days versus 7 days, P=0.03). The time from initiation of feedings to hospital discharge, which accounts for initiation age, significantly favored the exclusively HM-fed infants (median 7 days versus 10 days, P=0.01). CONCLUSION Exclusive HM feeding after gastroschisis repair decreases time to achieve full enteral feeds and time to discharge.
Collapse
Affiliation(s)
- J A Kohler
- Department of Pediatrics, Children's Hospital of The King's Daughters, Eastern Virginia Medical School, Norfolk, VA, USA.
| | | | | |
Collapse
|
29
|
Perrin MT, Fogleman A, Allen JC. The nutritive and immunoprotective quality of human milk beyond 1 year postpartum: are lactation-duration-based donor exclusions justified? J Hum Lact 2013; 29:341-9. [PMID: 23674288 DOI: 10.1177/0890334413487432] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Donor human milk is critical for the fragile preterm infant who does not have access to his or her mother's milk, improving survival rates and quality of survival and decreasing hospital stay. Despite the opening of donor milk banks around the world, shortages continue as demand for donor milk exceeds supply. One potential means of increasing supply is by reducing exclusion criteria that prohibit mothers from donating milk based on duration of lactation. Minimal research has been done on the composition of human milk during the second year of lactation, with most research focusing on the nutritive compounds and not the immunoprotective compounds. Several immunoprotective compounds, including lysozyme, lactoferrin, secretory immunoglobulin A, and oligosaccharides, are abundant in human milk compared to bovine-based infant formula and are partially or fully retained during Holder pasteurization, making them an important differentiating feature of donor milk. A PubMed search was conducted to review studies in human milk composition during the second year of lactation. Limitations of existing research include sample collection protocols, small study sizes, and use of populations that may have been at risk for nutritional deficiencies. Stable concentrations of several components were reported including protein, lactose, iron, copper, lactoferrin, and secretory immunoglobulin A. Lysozyme concentration increased during extended lactation, while zinc and calcium concentrations declined into the second year. Conflicting findings were reported on fat content, and no information was available regarding oligosaccharide content. More research is needed to create evidence-based guidelines regarding the nutritive and immunoprotective value of donor milk throughout the course of lactation.
Collapse
|
30
|
Tan-Dy CRY, Ohlsson A. Lactase treated feeds to promote growth and feeding tolerance in preterm infants. Cochrane Database Syst Rev 2013; 2013:CD004591. [PMID: 23543535 PMCID: PMC6984660 DOI: 10.1002/14651858.cd004591.pub3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Successful transition from parenteral nutrition to full enteral feedings during the immediate neonatal period is associated with improved growth in preterm infants. Lactase is the last of the major intestinal disaccharidases to develop in preterm infants. Because of inadequate lactase activity, preterm infants are unable to digest lactose. Lactase preparations could potentially be used to hydrolyse lactose in formulas and breast milk to minimize lactose malabsorption in preterm infants. OBJECTIVES To assess the effectiveness and safety of the addition of lactase to milk compared to placebo or no intervention for the promotion of growth and feeding tolerance in preterm infants. PRIMARY OUTCOMES weight gain expressed as grams/kg/day, growth expressed as weight, length and head circumference percentile for postmenstrual age (PMA), assessed at birth and at 40 weeks PMA, days to achieve full enteral feeds. SECONDARY OUTCOMES several common outcomes associated with preterm birth, and adverse effects. SEARCH METHODS Electronic and manual searches were conducted in January 2005 of the Cochrane Central Register of Controlled Trials (CENTRAL, The Cochrane Library, 2004, Issue 4), MEDLINE (1966 to Jan 2005), EMBASE (1980 to Jan 2005) and CINAHL (1982 to Jan 2005), personal files, bibliographies of identified trials and abstracts by the Pediatric Academic Societies' Meetings and the European Society of Pediatric Research Meetings published in Pediatric Research. The searches were repeated in May 2012 of The Cochrane Library, MEDLINE, EMBASE and CINAHL and abstracts from the Pediatric Academic Societies' Annual Meetings from 2000 to 2012 (Abstracts2View). The Web of Science was searched using the only previously identified trial by Erasmus 2002 as the starting point to search for additional trials that cited this trial. SELECTION CRITERIA Types of studies: randomized or quasi-randomized controlled trials. PARTICIPANTS preterm infants < 37 weeks PMA. INTERVENTION addition of lactase to milk versus placebo or no intervention. DATA COLLECTION AND ANALYSIS The standard methods of the Cochrane Neonatal Review Group were followed independently by the review authors to assess study quality and report outcomes. Treatment effects, calculated using Review Manager 5, included risk ratio (RR), risk difference (RD) and mean difference (MD), all with 95% confidence intervals (CI). A fixed-effect model was used for meta-analyses. We did not perform heterogeneity tests as only one study was identified. MAIN RESULTS The repeat searches conducted in May 2012 did not identify any additional studies for inclusion. One study enrolling 130 infants of 26 to 34 weeks PMA (mean postnatal age at entry 11 days) was identified and no identified study was excluded. The study was a double blind randomized controlled trial of high quality. Lactase treated feeds were initiated when enteral feedings provided > 75% of daily intake. None of the primary outcomes outlined in the protocol for this review and only one of the secondary outcomes, necrotizing enterocolitis (NEC) were reported on. The RR for NEC was 0.32 (95% CI 0.01 to 7.79); the RD was -0.02 (95% CI -0.06 to 0.03) (a reduction which was not statistically significant). There was a statistically significant increase in weight gain at study day 10 in the lactase treated feeds group but not at any other time points. Overall, there was not a statistically significant effect on weight gain. No adverse effects were noted. AUTHORS' CONCLUSIONS The only randomized trial to date provides no evidence of significant benefit to preterm infants from adding lactase to their feeds. Further research regarding effectiveness and safety are required before practice recommendations can be made. Randomized controlled trials comparing lactase versus placebo treated feeds and enrolling infants when enteral feeds are introduced are required. The primary and secondary outcomes for effectiveness and safety should include those identified in this review.
Collapse
|
31
|
Morgan J, Bombell S, McGuire W. Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants. Cochrane Database Syst Rev 2013; 2013:CD000504. [PMID: 23543508 PMCID: PMC11480887 DOI: 10.1002/14651858.cd000504.pub4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND The introduction of enteral feeds for very preterm (< 32 weeks) or very low birth weight (< 1500 grams) infants is often delayed due to concern that early introduction may not be tolerated and may increase the risk of necrotising enterocolitis. However, prolonged enteral fasting may diminish the functional adaptation of the immature gastrointestinal tract and extend the need for parenteral nutrition with its attendant infectious and metabolic risks. Trophic feeding, giving infants very small volumes of milk to promote intestinal maturation, may enhance feeding tolerance and decrease the time taken to reach full enteral feeding independently of parenteral nutrition. OBJECTIVES To determine the effect of early trophic feeding versus enteral fasting on feed tolerance, growth and development, and the incidence of neonatal morbidity (including necrotising enterocolitis and invasive infection) and mortality in very preterm or VLBW infants. SEARCH METHODS We used the standard search strategy of the Cochrane Neonatal Review Group. This included electronic searches of the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2012, Issue 12), MEDLINE, EMBASE and CINAHL (1980 until December 2012), conference proceedings and previous reviews. SELECTION CRITERIA Randomised or quasi-randomised controlled trials that assessed the effects of early trophic feeding (milk volumes up to 24 ml/kg/day introduced before 96 hours postnatal age and continued until at least one week after birth) versus a comparable period of enteral fasting in very preterm or very low birth weight infants. DATA COLLECTION AND ANALYSIS We extracted data using the standard methods of the Cochrane Neonatal Review Group with separate evaluation of trial quality and data extraction by two authors and synthesis of data using risk ratio, risk difference and mean difference. MAIN RESULTS Nine trials in which a total of 754 very preterm or very low birth weight infants participated were eligible for inclusion. Few participants were extremely preterm (< 28 weeks) or extremely low birth weight (< 1000 grams) or growth restricted. These trials did not provide any evidence that early trophic feeding affected feed tolerance or growth rates. Meta-analysis did not detect a statistically significant effect on the incidence of necrotising enterocolitis: typical risk ratio 1.07 (95% confidence interval 0.67 to 1.70); risk difference 0.01 (-0.03 to 0.05). AUTHORS' CONCLUSIONS The available trial data do not provide evidence of important beneficial or harmful effects of early trophic feeding for very preterm or very low birth weight infants. The applicability of these findings to extremely preterm, extremely low birth weight or growth restricted infants is limited. Further randomised controlled trials would be needed to determine how trophic feeding compared with enteral fasting affects important outcomes in this population.
Collapse
Affiliation(s)
- Jessie Morgan
- Hull York Medical School & Centre for Reviews and Dissemination, University of York, York, UK.
| | | | | |
Collapse
|
32
|
Abstract
We have used an expansive definition of a micropreterm infant as <30 weeks' gestation to provide a global perspective to a "high risk" group of preterm infants for which there are little published data to guide nutritional management. Consensus nutritional guidelines for preterm infants have been developed for infants >1000 g birth weight and >28 weeks' gestational age. Micropreterm infants have greater nutritional deficits at birth than more mature preterm infants and accumulate greater postnatal deficits. Nutritional guidelines based on the needs of preterm infants born >28 weeks' gestation are unlikely, on a theoretical basis, to meet nutritional requirements of micropreterm infants. Unfortunately, very few good quality studies have addressed the nutritional requirements of this group specifically; this makes it difficult to formulate solid, evidence-based nutritional recommendations for these neonates. Nutritional management of micropreterm infants is based on recommendations established for preterm infants, which are adjusted after considering an infant's gestational age, birth weight, and clinical status. Minimal enteral feeding should commence on the first or second day of life, with incremental advancement and fortification of human milk when 100 mL/kg is tolerated. Early use of parenteral nutrition is recommended, ideally initiated within the first hours of life and enteral feeds are being established; this will help prevent the accumulation of nutritional deficits and incidence of growth failure. Fortified human milk should be given in order to meet nutritional requirements. When human milk is not available in sufficient quantity, a preterm formula should be given.
Collapse
Affiliation(s)
- David Tudehope
- Mater Medical Research Institute and School of Medicine, The University of Queensland, Queensland, Australia.
| | | | | | | |
Collapse
|
33
|
Liao Y, Jiang R, Lönnerdal B. Biochemical and molecular impacts of lactoferrin on small intestinal growth and development during early life1This article is part of a Special Issue entitled Lactoferrin and has undergone the Journal's usual peer review process. Biochem Cell Biol 2012; 90:476-84. [DOI: 10.1139/o11-075] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Postnatal modeling of the intestinal epithelium has long-term impacts on the healthy development of infants and relies largely on nutrient composition of the diet. Lactoferrin (Lf) is among the various human milk trophic factors that facilitate the infant intestinal adaptation. Hydrolysis of Lf is minimal at the prevailing postprandial pH of infants, and Lf may therefore have greater biological potential in infants than in adults. Lf bidirectionally stimulates concentration-dependent proliferation and differentiation of small intestinal epithelial cells, and therefore affects small intestinal mass, length, and epithelial digestive enzyme expression. A 105 kDa Lf receptor (LfR) specifically mediates the uptake of Lf into enterocytes and crypt cells. Mechanistically, the complex of Lf and LfR is internalized through clathrin-mediated endocytosis; both iron-free apo-Lf and iron-saturated holo-Lf activate the PI3K/Akt pathway, whereas only apo-Lf triggers ERK1/2 signaling. Lf enters the nucleus, where it can stimulate thymidine incorporation into crypt cells, regulating transcription of genes such as TGF-β1. In the fetus, the plasma membrane LfR is at the highest abundance in the small intestine, and the receptor gene is tightly controlled at multiple levels. Aspecific microRNA, miR-584, is involved in the posttranscriptional regulation of LfR, and in the human LfR DNA promoter, 2 Sp1 binding sites have been characterized functionally. Finally, cell proliferation and global gene expression reveal that native bovine Lf can perform biological activities similar to those exerted by human Lf in postnatal small intestinal development.
Collapse
Affiliation(s)
- Yalin Liao
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Rulan Jiang
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
34
|
Kim JH, Froh EB. What Nurses Need To Know Regarding Nutritional and Immunobiological Properties of Human Milk. J Obstet Gynecol Neonatal Nurs 2012; 41:122-137. [DOI: 10.1111/j.1552-6909.2011.01314.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
35
|
Abstract
Feeding intolerance is a well-known phenomenon in the NICU and is linked to morbidity and mortality in the premature infant. However, a universal definition for this concept is lacking. Properly defining a concept is a key step in a successful research project. A concept analysis is an examination of the fundamental elements of a concept to bring clarification and definition to the topic of interest. The purpose of this concept analysis was to clarify the phenomenon of feeding intolerance in the premature infant and to provide a universal conceptual and operational definition for researchers and clinicians to use in practice theory.
Collapse
|
36
|
Braquehais FR, Cava MJB. Functionality of α-glucans in special formulas for infant and clinical nutrition. STARCH-STARKE 2011. [DOI: 10.1002/star.201000082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Cilieborg MS, Boye M, Thymann T, Jensen BB, Sangild PT. Diet‐Dependent Effects of Minimal Enteral Nutrition on Intestinal Function and Necrotizing Enterocolitis in Preterm Pigs. JPEN J Parenter Enteral Nutr 2011; 35:32-42. [DOI: 10.1177/0148607110377206] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Malene Skovsted Cilieborg
- Department of Human Nutrition, Faculty of Life Science, University of Copenhagen, Denmark
- National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Mette Boye
- National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Thomas Thymann
- Department of Human Nutrition, Faculty of Life Science, University of Copenhagen, Denmark
| | - Bent Borg Jensen
- Department of Animal Health and Bioscience, Faculty of Agricultural Sciences, University of Aarhus, Tjele, Denmark
| | - Per Torp Sangild
- Department of Human Nutrition, Faculty of Life Science, University of Copenhagen, Denmark
| |
Collapse
|
38
|
Shulman RJ, Ou CN, Smith EO. Evaluation of potential factors predicting attainment of full gavage feedings in preterm infants. Neonatology 2011; 99:38-44. [PMID: 20588069 PMCID: PMC3214900 DOI: 10.1159/000302020] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/09/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND The clinical measures of gastric residuals and abdominal distention are often used to guide feeding in preterm infants, but there are few data demonstrating their usefulness. Similarly, techniques are now available to investigate gastrointestinal (GI) function noninvasively and safely, but their ability to predict attainment of full gavage feedings and/or feeding volume in preterm infants is unclear. OBJECTIVE We sought to determine prospectively the potential relationships of attainment of full gavage feedings and feeding volume with clinical measures and noninvasive GI tests. METHODS Fifty preterm infants were followed prospectively. Daily tally was taken of gavage feeding intake, gastric residual volumes (GRVs; milliliters per day, number of GRVs >50% of the previous feeding volume, and number of GRVs >2 ml/kg), and abdominal distention. Infants underwent repeated measurement of lactase activity, GI permeability, fecal calprotectin concentration, and gastric emptying. RESULTS The number of GRVs >2 ml/kg tended to decrease with postnatal age (p = 0.06). Lactase activity and feeding volume in milliliters per kilogram per day prior to achieving full feedings were correlated (p = 0.007, β = 0.164). There was no correlation between feeding outcomes and GRV (ml/day), GRV >50%, GRV >2 ml/kg, small bowel, colonic, or whole bowel permeability, fecal calprotectin concentration, gastric emptying, or abdominal distention. CONCLUSIONS GRV is unreliable in predicting attainment of full gavage feeding. Lactase activity is related to feeding volume. However, other noninvasive GI tests utilized were not predictive. These data cast doubt upon the utility of GRV in guiding feeding therapy. Randomized trials of different GRV management protocols are needed.
Collapse
Affiliation(s)
- Robert J Shulman
- Department of Pediatrics, Children's Nutrition Research Center, Houston, Tex 77030-2600, USA. rshulman @ bcm.tmc.edu
| | | | | |
Collapse
|
39
|
Sisk P, Quandt S, Parson N, Tucker J. Breast milk expression and maintenance in mothers of very low birth weight infants: supports and barriers. J Hum Lact 2010; 26:368-75. [PMID: 20930219 DOI: 10.1177/0890334410371211] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The study objective was to identify patterns of factors that supported or hindered initiation of breast milk expression and maintenance of breast milk production after the birth of a very low birth weight (VLBW) infant in a sample of US women with varied prenatal infant feeding intentions. In-depth interviews were conducted 1 to 6 months after delivery in 32 women who initiated breast milk expression after encouragement from hospital staff. Pregnancy complications, anxiety regarding their infant's health, and lack of privacy interfered with initiation of milk expression. After hospital discharge, using manual or small electric breast pumps, travel to the neonatal intensive care unit, return to work, and difficulty with time management interfered with maintenance of breast milk production. Family support, positive attitudes toward pumping, and anticipation of breastfeeding supported maintenance of breast milk production. From these data emerge points of intervention where additional support could improve mothers' experiences and increase duration of breast milk feeding.
Collapse
Affiliation(s)
- Paula Sisk
- Department of Pediatrics, Wake Forest University Health Sciences, Sara Lee Center for Women's Health, Neonatal Intensive Care Unit, Winston-Salem, North Carolina 27103, USA
| | | | | | | |
Collapse
|
40
|
Sisk PM, Lovelady CA, Dillard RG, Gruber KJ, O'Shea TM. Maternal and infant characteristics associated with human milk feeding in very low birth weight infants. J Hum Lact 2009; 25:412-9. [PMID: 19602575 DOI: 10.1177/0890334409340776] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study identified maternal and infant characteristics predicting human milk (HM) feeding in very low birth weight (VLBW) infants whose mothers (n = 184) participated in a study of lactation counseling and initiated milk expression. Data were collected prospectively, by maternal interview and medical record review. During hospitalization, 159 (86%) infants received at least 50% HM proportion of feedings in the first 2 weeks of life, and 114 (62%) received some HM until the day of hospital discharge. Analysis showed plan to breastfeed was the strongest predictor of initiation and duration of HM feeding. Greater than 12 years of education, respiratory distress syndrome, Apgar score >6, and female gender were significant predictors, and no perinatal hypertensive disorder, white race, and mechanical ventilation were marginal predictors of HM feeding. Women with a high-risk pregnancy should be provided education about the benefits of breastfeeding for infants who are likely to be born prematurely.
Collapse
Affiliation(s)
- Paula M Sisk
- Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
BACKGROUND The introduction of enteral feeds for very low birth weight (VLBW) infants is often delayed due to concern that early introduction may not be tolerated and may increase the risk of necrotising enterocolitis. However, enteral fasting may diminish the functional adaptation of the immature gastrointestinal tract and prolong the need for parenteral nutrition with its attendant infectious and metabolic risks. Early trophic feeding, giving infants very small volumes of milk during the first week after birth, may promote intestinal maturation, enhance feeding tolerance and decrease time to reach full enteral feeding independently of parenteral nutrition. OBJECTIVES To determine the effect of early trophic feeding versus enteral fasting on feed tolerance, growth, and the incidence of necrotising enterocolitis, mortality and other morbidities in VLBW infants. SEARCH STRATEGY The standard search strategy of the Cochrane Neonatal Group was used. Searches were made of the Cochrane Central Register of Controlled Trials (CENTRAL, The Cochrane Library, Issue 1, 2009), MEDLINE (1966 - February 2009), EMBASE (1980 - February 2009), CINAHL (1982 - February 2009), conference proceedings, and previous reviews. SELECTION CRITERIA Randomised or quasi-randomised controlled trials that assessed the effects of early trophic feeding (milk volumes up to 24 ml/kg/day introduced before 96 hours postnatal age and continued until at least one week after birth) versus a comparable period of enteral fasting in VLBW infants. DATA COLLECTION AND ANALYSIS The standard methods of the Cochrane Neonatal Group were used, with separate evaluation of trial quality and data extraction by two review authors. Data were synthesised using a fixed effects model and reported using typical relative risk, typical risk difference and weighted mean difference. MAIN RESULTS Nine trials, in which a total of 754 VLBW infants participated, were eligible for inclusion. These trials did not provide any evidence that early trophic feeding affected feed tolerance or growth rates in VLBW infants. Meta-analysis did not detect a statistically significant effect on the incidence of necrotising enterocolitis: typical relative risk 1.07 (95% confidence interval 0.67, 1.70); typical risk difference 0.01 (95% confidence interval -0.04, 0.05). AUTHORS' CONCLUSIONS The available data cannot exclude important beneficial or harmful effects and are insufficient to inform clinical practice. Further large pragmatic randomised controlled trials are needed to determine how early trophic feeding compared with enteral fasting affects important clinical outcomes in VLBW infants.
Collapse
Affiliation(s)
- Sarah Bombell
- Centre for Newborn Care, Australian National University, Canberra Hospital, Canberra, Australia, ACT 2606
| | | |
Collapse
|
42
|
Sisk PM, Lovelady CA, Gruber KJ, Dillard RG, O'Shea TM. Human milk consumption and full enteral feeding among infants who weigh </= 1250 grams. Pediatrics 2008; 121:e1528-33. [PMID: 18519456 DOI: 10.1542/peds.2007-2110] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Establishing enteral feeding is an important goal in the care of very low birth weight infants. In such infants, receipt of >/=50 mL/kg per day human milk during hospitalization has been associated with shorter time to full enteral feeding. The objective of this study was to determine whether high proportions (>/=50%) of human milk during feeding advancement are associated with shorter time to full enteral feeding and improved feeding tolerance. METHODS This was a prospective cohort study of very low birth weight infants (n = 127) who were grouped into low (<50%; n = 34) and high (>/=50%; n = 93) human milk consumption groups according to their human milk proportion of enteral feeding during the time of feeding advancement. The primary outcomes of interest were ages at which 100 and 150 mL/kg per day enteral feedings were achieved. RESULTS The high human milk group reached 100 mL/kg per day enteral feeding 4.5 days faster than the low human milk group. The high human milk group reached 150 mL/kg per day enteral feeding 5 days faster than the low human milk group. After adjustment for gestational age, gender, and respiratory distress syndrome, times to reach 100 and 150 mL/kg per day were significantly shorter for those in the high human milk group. Infants in the high human milk group had a greater number of stools per day; other indicators of feeding tolerance were not statistically different. CONCLUSION In infants who weighed </=1250 g, enteral feeding that contained at least 50% maternal human milk was associated with fewer days to full enteral feedings.
Collapse
Affiliation(s)
- Paula M Sisk
- Department of Pediatrics, Wake Forest University School of Medicine, One Medical Center Blvd, Winston-Salem, NC 27157, USA.
| | | | | | | | | |
Collapse
|
43
|
Effect of enteral IGF-1 supplementation on feeding tolerance, growth, and gut permeability in enterally fed premature neonates. J Pediatr Gastroenterol Nutr 2008; 46:184-90. [PMID: 18223378 DOI: 10.1097/mpg.0b013e31815affec] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The gastrointestinal tract of the premature newborn functions suboptimally with regard to digestion, absorption, and feeding tolerance. Human milk contains trophic factors, such as insulin-like growth factor-1 (IGF-1), that are believed to stimulate gut growth and function. The objective of this double blind, randomized, controlled trial was to assess the effects of enteral IGF-1 supplementation on whole body growth measured by weight gain (in grams per kilogram per day), days to regain birth weight, and anthropometrical characteristics, and gut maturation and permeability (measured by sugar absorption tests). PATIENTS AND METHODS The study included 60 premature infants (birth weight 750-1250 g) during the first month of life. Patients received either standard infant formula or standard infant formula supplemented with IGF-1 in a concentration twice that of human colostrum (10 microg/100 mL of formula). Primary endpoints were days to full enteral feeding, days to regain birth weight, and growth rate. Sugar absorption tests were performed weekly to assess the secondary endpoints gut permeability and maturation. RESULTS None of the primary endpoints differed to statistical significance between groups at any point. However, gut permeability was significantly lower in the IGF-1 supplement group on day 14 compared with the control group. At day 21, lactulose/mannitol excretion ratios were (again) comparable between the groups. CONCLUSIONS Although gut permeability showed a faster decrease in the IGF-1 supplement group, our data do not support IGF-1 supplementation to infant formula.
Collapse
|
44
|
Yang Q, Smith PB, Goldberg RN, Cotten CM. Dynamic change of fecal calprotectin in very low birth weight infants during the first month of life. Neonatology 2008; 94:267-71. [PMID: 18784422 PMCID: PMC2790758 DOI: 10.1159/000151645] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Accepted: 11/13/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Calprotectin is a cytosolic component of neutrophils. Fecal calprotectin (FC) level is a useful marker for exacerbation of inflammatory bowel disease in children. FC may be a useful marker for necrotizing enterocolitis (NEC). OBJECTIVE To determine normal baseline levels of FC and observe dynamic changes of FC levels over the first postnatal month in very low birth weight (VLBW) infants. METHODS FC levels of 14 VLBW infants (gestational age 23-30 weeks, birth weight <or=1,500 g) were serially measured in the first postnatal month. Demographics, feeding regimens, antibiotic use, laboratory and x-ray results, and maternal information were recorded. We assessed how FC levels changed over time, varied with nutritional source and differed between sick versus well infants. RESULTS FC levels were not related to gestational age or feedings regimen. FC levels tended to decrease with increasing age (p = 0.121) and feeding volumes (p = 0.179). FC levels differed between 'well' and 'sick' infants (122.8 +/- 98.9 vs. 380.4 +/- 246.3 microg/g stool, p < 0.001). FC >350 microg/g stool was noted with signs of gastrointestinal injury, such as bloody stool and bowel perforation. FC levels decreased after initiation of treatments in sick infants who recovered. CONCLUSIONS FC levels may be a marker for early diagnosis and resolution of gastrointestinal illnesses in VLBW infants. Its utility for early diagnosis and assessment of resolution of NEC should be studied in a larger cohort of VLBW infants.
Collapse
Affiliation(s)
- Qing Yang
- Department of Pediatrics, Division of Neonatology, Duke University Medical Center, Durham, NC, USA.
| | | | | | | |
Collapse
|
45
|
Abstract
The level of gastrointestinal (GI) maturity of an individual infant is a major determinant of whether the infant will be able to meet nutritional needs by sole use of the GI tract or if parenteral means will be necessary. The GI tract is not only an organ for digestion and absorption of nutrients; it also performs major endocrine, neural and immunologic functions. In this review, anatomic, functional and biochemical development will be described and related to means by which enteral nutrition can be used in the prematurely born infant to optimize health and prevent disease.
Collapse
Affiliation(s)
- Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, USA.
| |
Collapse
|
46
|
Meloncelli PJ, Williams TM, Hartmann PE, Stick RV. The synthesis of 2-, 3-, 4- and 6-O-α-d-glucopyranosyl-d-galactopyranose, and their evaluation as nutritional supplements for pre-term infants. Carbohydr Res 2007; 342:1793-804. [PMID: 17517382 DOI: 10.1016/j.carres.2007.04.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 04/20/2007] [Accepted: 04/25/2007] [Indexed: 11/25/2022]
Abstract
Four methods have been screened for the synthesis of some alpha-D-glucopyranosides, with the recently reported (Mukaiyama) combination of 2,3,4,6-tetra-O-benzyl-alpha-D-glucopyranosyl iodide and triphenylphosphine oxide being the most successful, especially in the diastereoselectivity exhibited. The alpha-D-glucopyranosides so obtained have been deprotected to yield 2-, 3-, 4- and 6-O-alpha-D-glucopyranosyl-D-galactopyranose. Only the last disaccharide showed any hydrolysis by alpha-glycosidases but this success was not emulated by mucosal extracts from the small intestine of the pig.
Collapse
Affiliation(s)
- Peter J Meloncelli
- Chemistry M313, School of Biomedical, Biomolecular and Chemical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | | | | | | |
Collapse
|
47
|
Commare CE, Tappenden KA. Development of the infant intestine: implications for nutrition support. Nutr Clin Pract 2007; 22:159-73. [PMID: 17374790 DOI: 10.1177/0115426507022002159] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The incidence of preterm births has continued to increase over the past 25 years, and therefore the optimal feeding of these infants is an important clinical concern. This review focuses on intestinal development and physiology, with a particular emphasis on developmentally immature functions of the preterm intestine and the resulting implications for nutrition therapies used to feed the preterm infant.
Collapse
Affiliation(s)
- Coryn E Commare
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | |
Collapse
|
48
|
Abstract
Providing optimal nutrition to satisfy the growth needs of very low birth weight infants is critical. The available preterm formulas and fortified human milk diets provide protein intakes of approximately 3.5 to 3.6 g/kg/d when volumes sufficient to provide 120 kcal/kg/d are fed to these infants. These intakes support growth and protein accretion at about or slightly greater than intrauterine rate and lead to relatively increased fat deposition. However, most very low birth infants fed these diets remain below the 10th percentile of the intrauterine growth standards at discharge. There is clear evidence that, with respect to growth, very low birth infants are likely to benefit from a higher protein intake; however, there is no clear evidence that energy intakes greater than 120 kcal/kg/d are needed. Although the upper limit of protein intake and the ideal protein:energy ratio remain controversial, there is enough evidence to support the beneficial and safe use of formulas providing protein:energy ratio of 3.2 to 3.3 g/100 kcal.
Collapse
Affiliation(s)
- Sudha Kashyap
- Division of Neonatalogy, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
49
|
Buccigrossi V, de Marco G, Bruzzese E, Ombrato L, Bracale I, Polito G, Guarino A. Lactoferrin induces concentration-dependent functional modulation of intestinal proliferation and differentiation. Pediatr Res 2007; 61:410-4. [PMID: 17515863 DOI: 10.1203/pdr.0b013e3180332c8d] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Human milk stimulates intestinal development through the effects of various moieties. Lactoferrin (LF) is a glycoprotein of human milk whose concentration is highest in colostrum decreasing in mature milk. LF promotes enterocyte growth in intestinal cell lines. We tested the hypothesis that LF induces a distinct effect on enterocyte proliferation and differentiation, depending on its concentration. We examined the dose-related effects by human-native LF (N-LF) in Caco-2 (human colon adenocarcinoma) cells. At high concentrations, N-LF stimulated cell proliferation in immature Caco-2 cells, as judged by 3H-thymidine incorporation. In contrast, sucrase and lactase activities were increased at low but not high LF concentrations and their mRNA were also increased, indicating a transcriptional effect. Because iron binds specific LF sites, we compared the potency of N-LF and iron-saturated LF (I-LF) and found the native form more potent. Finally, we tested the effects by bovine LF (bLF) in the same system and found the latter more potent than the human isoform in inducing cell growth and lactase expression. These results suggest that LF directly induces enterocyte growth and proliferation, depending on its concentration, thereby regulating the earlyx postnatal intestinal development. bLF could be added to infant formula as a growth factor in selected intestinal diseases.
Collapse
|
50
|
Street JL, Montgomery D, Alder SC, Lambert DK, Gerstmann DR, Christensen RD. Implementing feeding guidelines for NICU patients<2000 g results in less variability in nutrition outcomes. JPEN J Parenter Enteral Nutr 2007; 30:515-8. [PMID: 17047177 DOI: 10.1177/0148607106030006515] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND We devised a consistent approach to instituting and advancing enteral nutrition among neonatal intensive care unit (NICU) patients<2000 g birth weight. We then assessed variability in feeding-related outcomes during a period before (period 1) vs after (period 2) implementing these guidelines. METHODS Using data from period 1 vs period 2, we statistically compared the equivalence of variance, focusing on certain feeding-related outcomes. Specific outcomes we chose to examine were (1) day of life when the first enteral feedings were given, (2) number of days during the entire hospitalization when no feedings were given, (3) number of days parenteral nutrition (PN) was administered, and (4) day of life when feedings of 80 mL/k/d and 100 kcal/k/d enteral were achieved. RESULTS Fifty-eight patients<2000 g were admitted to the NICU in period 1, of which 56 survived to discharge home. In period 2, 68 patients<2000 g were admitted and 66 survived to discharge. Demographic features of the patients in periods 1 and 2 did not differ. In both periods, feedings were begun on a median of day 1. However, in period 1 the range was from day 0 to day 24, and in period 2, the range was from day 0 to day 6 (equivalence of variance p<.001). After feedings were initiated, they were withheld for a median of 2 days (range, 0-23) during the remainder of the hospitalization in period 1 vs a median of 1 day (range, 0-12) in period 2 (p<.001). During period 1, PN was used for a median of 10 days (range, 0-72) vs 7 (range, 0-47) in period 2 (p=.001). During period 1, more variability occurred in the day of life when 80 mL/k/d and 100 kcal/k/d were achieved (both p<.001). No differences were seen in necrotizing enterocolitis, intestinal perforation, mortality, or length of hospital stay. CONCLUSIONS Implementing feeding guidelines was associated with significantly less variability in feeding-related outcomes. We speculate that this is a reflection of better feeding tolerance, which resulted from a more consistent approach to initiating and advancing enteral feedings.
Collapse
Affiliation(s)
- Jennifer L Street
- Intermountain Healthcare Neonatology Research, McKay-Dee Hospital Center, Ogden 84403, and Department of Family and Preventive Medicine, University of Utah School of Medicine, Salt Lake City, USA
| | | | | | | | | | | |
Collapse
|