1
|
Puri K, Svenstrup C, Vanderpool C. Functional Infant Formula Additives. Neoreviews 2025; 26:e163-e171. [PMID: 40020744 DOI: 10.1542/neo.26-3-020] [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: 08/30/2025] [Accepted: 11/04/2025] [Indexed: 03/03/2025]
Abstract
Breastfeeding is the ideal initial feeding method for providing nutrition to full-term infants and is recommended by major health organizations, including the American Academy of Pediatrics and the World Health Organization. Despite improvements in global breastfeeding rates, many infants still receive formula. Significant advancements have been achieved in the safety and nutritional content of modern formulas. Various functional additives, such as human milk oligosaccharides, milk fat globule membrane, docosahexaenoic acid, and lactoferrin, are used with the aim to replicate some of the benefits of breast milk. These additives enhance formula by providing benefits beyond basic nutrition. The aim of this review is to summarize these additives and their impact on infant nutrition and development.
Collapse
Affiliation(s)
- Kanika Puri
- Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana
| | - Courtney Svenstrup
- Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana
| | - Charles Vanderpool
- Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana
| |
Collapse
|
2
|
Witte Castro A, Couce ML, de Lamas C, López-Giménez MR, Jiménez Varas MÁ, Zozaya C, Saenz de Pipaon M. Long-chain polyunsaturated fatty acids supplementation and sepsis: a systematic review and meta-analysis. Pediatr Res 2025; 97:924-938. [PMID: 39300278 DOI: 10.1038/s41390-024-03579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/30/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Long chain polyunsaturated fatty acids (LCPUFAs) have proven to be essential for development in preterm infants and have been studied for their capacity to reduce inflammation and infection rates, including sepsis in enteral and parenteral nutrition. The aim of this review and meta-analysis is to gather the information available on this subject to determine if n-3 polyunsaturated fatty acids can reduce sepsis incidence in preterm infants. METHODS This systematic review was conducted by searching in the databases MEDLINE (via PubMed), ISI-Web of Science, EMBASE, SCOPUS, SciELO, and Cochrane Library databases. We analyzed the data regarding sepsis using the Grading of Recommendations Assessment, Development and Evaluation approach to assess the quality of the evidence. RESULTS A total of 40 trials were included for review and 35 trials had the data available for quantitative analysis. LCPUFAs supplementation did not reduce incidence of sepsis (relative risk (RR), confidence interval (CI) 0.95 [0.87, 1.03] P = 0.87; I2 = 0%). These results remained consistent after the sensitivity analysis. CONCLUSION The results of this systematic review and meta-analysis indicate that LCPUFA supplementation is not associated with a significant decrease in the incidence of sepsis in premature infants. IMPACT Reviewing the information available about LCPUFA supplementation and sepsis since the results in previous Clinical Trials (CT) are inconclusive. It summarizes the results of 42 CT and we have not found conclusive results regarding sepsis in the literature. It could be of clinical interest for pediatricians and nutritionists.
Collapse
Affiliation(s)
| | - María L Couce
- Department of Forensic Sciences, Pathological Anatomy, Gynecology and Obstetrics and Pediatrics, University of Santiago de Compostela, Santiago de Compostela, Spain
- Division of Neonatology, University Clinical Hospital of Santiago de Compostela, IDIS-Sanitary Research Institute of Santiago de Compostela, RICORS-SAMID, CIBERER, Santiago de Compostela, Spain
| | - Carmela de Lamas
- Department of Forensic Sciences, Pathological Anatomy, Gynecology and Obstetrics and Pediatrics, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | - C Zozaya
- Neonatology, Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ (La Paz University Hospital-Universidad Autónoma de Madrid), Madrid, Spain
| | - Miguel Saenz de Pipaon
- Neonatology, Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ (La Paz University Hospital-Universidad Autónoma de Madrid), Madrid, Spain
| |
Collapse
|
3
|
Drenckpohl DC, Christifano DN, Carlson SE. Is choline deficiency an unrecognized factor in necrotizing enterocolitis of preterm infants? Pediatr Res 2024; 96:875-883. [PMID: 38658665 DOI: 10.1038/s41390-024-03212-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/23/2024] [Accepted: 03/15/2024] [Indexed: 04/26/2024]
Abstract
We undertook this review to determine if it is plausible that choline or phosphatidylcholine (PC) deficiency is a factor in necrotizing enterocolitis (NEC) after two clinical trials found a dramatic and unexpected reduction in NEC in an experimental group provided higher PC compared to a control group. Sources and amounts of choline/PC for preterm infants are compared to the choline status of preterm infants at birth and following conventional nutritional management. The roles of choline/PC in intestinal structure, mucus, mesenteric blood flow, and the cholinergic anti-inflammatory system are summarized. Low choline/PC status is linked to prematurity/immaturity, parenteral and enteral feeding, microbial dysbiosis and hypoxia/ischemia, factors long associated with the risk of developing NEC. We conclude that low choline status exists in preterm infants provided conventional parenteral and enteral nutritional management, and that it is plausible low choline/PC status adversely affects intestinal function to set up the vicious cycle of inflammation, loss of intestinal barrier function and worsening tissue hypoxia that occurs with NEC. In conclusion, this review supports the need for randomized clinical trials to test the hypothesis that additional choline or PC provided parenterally or enterally can reduce the incidence of NEC in preterm infants. IMPACT STATEMENT: Low choline status in preterm infants who are managed by conventional nutrition is plausibly linked to the risk of developing necrotizing enterocolitis.
Collapse
Affiliation(s)
- Douglas C Drenckpohl
- Department of Food & Nutrition, OSF Healthcare Saint Francis Medical Center, Peoria, IL, 61637, USA
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, 66106, USA
| | - Danielle N Christifano
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, 66106, USA
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, 66106, USA.
| |
Collapse
|
4
|
Yao D, Ranadheera CS, Shen C, Wei W, Cheong LZ. Milk fat globule membrane: composition, production and its potential as encapsulant for bioactives and probiotics. Crit Rev Food Sci Nutr 2023; 64:12336-12351. [PMID: 37632418 DOI: 10.1080/10408398.2023.2249992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2023]
Abstract
Milk fat globule membrane (MFGM) is a complex trilayer structure present in mammalian milk and is mainly composed of phospholipids and proteins (>90%). Many studies revealed MFGM has positive effects on the immune system, brain development, and cognitive function of infants. Probiotics are live microorganisms that have been found to improve mental health and insulin sensitivity, regulate immunity, and prevent allergies. Probiotics are unstable and prone to degradation by environmental, processing, and storage conditions. In this review, the processes used for encapsulation of probiotics particularly the potential of MFGM and its constituents as encapsulating materials for probiotics are described. This study analyzes the importance of MFGM in encapsulating bioactive substances and emphasizes the interaction with probiotics and the gut as well as its resistance to adverse environmental factors in the digestive system when used as a probiotic embedding material. MFGM can enhance the gastric acid resistance and bile resistance of probiotics, mainly manifested in the survival rate of probiotics. Due to the role of digestion, MFGM-coated probiotics can be released in the intestine, and due to the biocompatibility of the membrane, it can promote the binding of probiotics to intestinal epithelial cells, and promote the colonization of some probiotics in the intestine.
Collapse
Affiliation(s)
- Dan Yao
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Science, Ningbo University, Ningbo, China
| | - Chaminda Senaka Ranadheera
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, Melbourne, Victoria, Australia
| | - Cai Shen
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, Melbourne, Victoria, Australia
- China Beacons Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Wei Wei
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ling-Zhi Cheong
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Alshaikh BN, Reyes Loredo A, Yusuf K, Maarouf A, Fenton TR, Momin S. Enteral long-chain polyunsaturated fatty acids and necrotizing enterocolitis: A systematic review and meta-analysis. Am J Clin Nutr 2023; 117:918-929. [PMID: 37137615 DOI: 10.1016/j.ajcnut.2023.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Preterm infants are at risk of long-chain polyunsaturated fatty acid (LCPUFA) deficiency. Recent studies on high-dose DHA; n-3 LCPUFA in preterm infants suggested potential positive effects on cognitive outcomes but raised concerns about some increased neonatal morbidities. These studies and recent recommendations for DHA supplementation generated controversy owing to the lack of balance between DHA and arachidonic acid (ARA; n-6 LCPUFA). OBJECTIVES To identify the effect of enteral supplementation of DHA, with and without ARA, on necrotizing enterocolitis (NEC) in very preterm infants. METHODS A systematic review of randomized and controlled trials compared enteral LCPUFAs with placebo or no supplementation in very preterm infants. We searched PubMed, Ovid-MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, and CINHAL databases from inception to July 2022. Data were extracted in duplicate using a structured proforma. A meta-analysis and metaregression with random-effects models were used. The interventions evaluated were DHA alone vs. that combined with ARA, source of DHA, dose, and supplement delivery methods. Methodological qualities and risk of bias were assessed using the Cochrane risk-of-bias tool. RESULTS Fifteen randomized clinical trials (RCTs) included 3963 very preterm infants with 217 cases of NEC. Supplementation with DHA alone increased NEC (2620 infants; RR: 1.56; 95% CI: 1.02, 2.39) with no evidence of heterogeneity (I2 = 0.0%, P = 0.46). Multiple metaregression revealed significant reduction in NEC when ARA was supplemented with DHA (aRR 0.42; 95% CI: 0.21, 0.88). The source of DHA, dose, and feeding type revealed no associations with NEC. Two RCTs supplemented high-dose DHA to lactating mothers. There was a significant increase in risk of NEC with this approach (1148 infants; RR: 1.92; 95% CI: 1.02, 3.61) with no evidence of heterogeneity (I2 = 0.0, P = 0.81). CONCLUSIONS Supplementation with DHA alone may increase risk of NEC. Concurrent supplementation with ARA needs to be considered when adding DHA to preterm infants' diet.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Nutrition and Gastroenterology Program, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.
| | - Adriana Reyes Loredo
- Neonatal Nutrition and Gastroenterology Program, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Kamran Yusuf
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Ahmed Maarouf
- Neonatal Nutrition and Gastroenterology Program, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Tanis R Fenton
- Community Health Sciences, Institute of Public Health, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Nutrition Services, Alberta Health Services, Calgary AB, Canada
| | - Sarfaraz Momin
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| |
Collapse
|
6
|
Abstract
Fatty acids are critical bioactives for fetal and neonatal development. Premature delivery and current nutritional strategies pose several challenges in restoring fatty acid balance in the preterm infant. The impact on fatty acid balance and outcomes using lipid emulsions, enteral nutrition, and enteral supplements are reviewed, including a summary of the most recent large clinical trials of enteral fatty acid supplementation for the preterm infant. Research gaps remain in successfully implementing nutritional strategies to optimize fatty acid status in preterm infants.
Collapse
|
7
|
Alshaikh BN, Reyes Loredo A, Knauff M, Momin S, Moossavi S. The Role of Dietary Fats in the Development and Prevention of Necrotizing Enterocolitis. Nutrients 2021; 14:145. [PMID: 35011027 PMCID: PMC8746672 DOI: 10.3390/nu14010145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 11/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of mortality and morbidity in preterm infants. The pathogenesis of NEC is not completely understood; however, intestinal immaturity and excessive immunoreactivity of intestinal mucosa to intraluminal microbes and nutrients appear to have critical roles. Dietary fats are not only the main source of energy for preterm infants, but also exert potent effects on intestinal development, intestinal microbial colonization, immune function, and inflammatory response. Preterm infants have a relatively low capacity to digest and absorb triglyceride fat. Fat may thereby accumulate in the ileum and contribute to the development of NEC by inducing oxidative stress and inflammation. Some fat components, such as long-chain polyunsaturated fatty acids (LC-PUFAs), also exert immunomodulatory roles during the early postnatal period when the immune system is rapidly developing. LC-PUFAs may have the ability to modulate the inflammatory process of NEC, particularly when the balance between n3 and n6 LC-PUFAs derivatives is maintained. Supplementation with n3 LC-PUFAs alone may have limited effect on NEC prevention. In this review, we describe how various fatty acids play different roles in the pathogenesis of NEC in preterm infants.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Adriana Reyes Loredo
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Megan Knauff
- Nutrition Services, Alberta Health Services, Calgary, AB T2N 2T9, Canada
| | - Sarfaraz Momin
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Shirin Moossavi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
- International Microbiome Centre, Cumming School of Medicine, Health Sciences Centre, University of Calgary, Calgary, AB T2N 2T9, Canada
| |
Collapse
|
8
|
Abou El Fadl DK, Ahmed MA, Aly YAF, Darweesh EAG, Sabri NA. Impact of Docosahexaenoic acid supplementation on proinflammatory cytokines release and the development of Necrotizing enterocolitis in preterm Neonates: A randomized controlled study. Saudi Pharm J 2021; 29:1314-1322. [PMID: 34819793 PMCID: PMC8596146 DOI: 10.1016/j.jsps.2021.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/14/2021] [Indexed: 10/26/2022] Open
Abstract
INTRODUCTION Preterm neonates have under-developed immune-regulatory system; consequently, there is a risk for developing chronic inflammation. Necrotizing enterocolitis (NEC) is an acute devastating neonatal intestinal inflammatory disorder. Due to the obscure multifactorial etiology, early diagnosis and effective treatment of NEC are limited. Consequently, effective strategies in the prevention of NEC, including nutritional approaches, are critically needed. The current study was conducted to assess the potential immunomodulatory effect of Docosahexaenoic Acid (DHA) supplementation in preterm neonates at neonatal intensive care unit (NICU) and subsequently its effect on preventing or reducing NEC incidence. METHODS This was a prospective randomized controlled study. A total of 67 neonates, with gestational age equal or less than 32 weeks at birth and weight less than or equal 1500 g, were randomly assigned to either DHA group or the control group. Modified Bell's staging criteria for NEC was used as an objective tool for diagnosis and staging of NEC. Levels of Interleukin 1 beta (IL-1β) were measured at baseline and after 10 days. Mortality and NICU length of stay (LOS) were also monitored. RESULTS Thirty neonates of each group completed the study. A statistically significant difference was observed between the two groups regarding diagnosis and staging of NEC (p = 0.0001). There was also a statistically significant difference between DHA group 22(73.3), 95% CI [55.9, 86.5] and the control group 8 (26.7), 95% CI [13.5, 44.1] in the percentage change in IL-1β levels (p = 0.0001).A statistically significant association was found between IL and 1 β change and NEC diagnosis (p = 0.001). NICU LOS was significantly lower among DHA group 21.63 ± 6.67 compared to the control group 25.07 ± 4.67 (p = 0.025). Mortality n (%) among the control group 4 (11.8) was higher than DHA group 3 (9.1), however, no significant difference was detected (p = 1.0). CONCLUSION Findings of this study suggest that enteral DHA supplementation can reduce NEC incidence in preterm neonates through its immunoregulatory effect that modulates production of regulatory cytokines.Trial registration: Registered at clinical trials.gov (NCT03700957), 6 October 2018.
Collapse
Affiliation(s)
- Dina Khaled Abou El Fadl
- Pharmacy Practice & Clinical Pharmacy Department, Faculty of Pharmaceutical Sciences & Pharmaceutical Industries, Future University in Egypt, Cairo, Egypt
| | - Marwa Adel Ahmed
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Yasmin AF Aly
- Paediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ebtissam Abdel Ghaffar Darweesh
- Pharmacy Practice & Clinical Pharmacy Department, Faculty of Pharmaceutical Sciences & Pharmaceutical Industries, Future University in Egypt, Cairo, Egypt
| | - Nagwa A. Sabri
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Bernabe-García M, Calder PC, Villegas-Silva R, Rodríguez-Cruz M, Chávez-Sánchez L, Cruz-Reynoso L, Mateos-Sánchez L, Lara-Flores G, Aguilera-Joaquín AR, Sánchez-García L. Efficacy of Docosahexaenoic Acid for the Prevention of Necrotizing Enterocolitis in Preterm Infants: A Randomized Clinical Trial. Nutrients 2021; 13:nu13020648. [PMID: 33671220 PMCID: PMC7922869 DOI: 10.3390/nu13020648] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 01/10/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel disease and a leading cause of morbidity and mortality in preterm infants. In this study, a randomized double-blind parallel-group (1:1) trial was carried out in two neonatal intensive care units of two tertiary hospitals. Two hundred and twenty-five preterm newborns with an expected functional gastrointestinal tract were recruited and received an enteral dose of 75 mg of docosahexaenoic acid (DHA)/kg body weight or high-oleic sunflower oil daily for 14 days from the first enteral feed after birth. Confirmed NEC was evaluated with Bell’s scale from stage ≥ IIa. Two hundred and fourteen randomized infants were analyzed in terms of the intent-to-treat (DHA-group: n = 105; control-group: n = 109); data for two hundred infants were analysed per protocol. Confirmed NEC was lower in infants from the DHA-group compared with the control-group (0/100 vs. 7/100; p = 0.007), with RR = 0.93 (95% CI 0.881 to 0.981), risk difference = −7%, (95% CI −12.00 to −1.99), and number needed-to-treat = 15 (95% CI 8.3 to 50). Intent-to-treat analysis showed a lower level of treatment failure in the DHA-group compared with the control-group (6/105 (6%) vs. 16/109 (15%); p = 0.03, RR = 0.905, (95% CI 0.826 to 0.991)). The results after multivariate-regression analysis remained significant. Adverse events (apart from the incidence of NEC) were not different between groups. A daily dose of DHA for 14 days starting with the first enteral feed may prevent NEC in preterm infants.
Collapse
Affiliation(s)
- Mariela Bernabe-García
- Unidad de Investigación Médica en Nutrición, UMAE Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México City 06720, Mexico; (M.B.-G.); (M.R.-C.)
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
- Correspondence:
| | - Raúl Villegas-Silva
- Neonatología, Hospital Infantil de México Federico Gómez, México City 06720, Mexico;
| | - Maricela Rodríguez-Cruz
- Unidad de Investigación Médica en Nutrición, UMAE Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México City 06720, Mexico; (M.B.-G.); (M.R.-C.)
| | - Luis Chávez-Sánchez
- Unidad de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México City 06720, Mexico;
| | - Leonardo Cruz-Reynoso
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia No.3, CMN La Raza, Instituto Mexicano del Seguro Social, México City 02990, Mexico; (L.C.-R.); (A.R.A.-J.); (L.S.-G.)
| | - Leovigildo Mateos-Sánchez
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia N° 4 “Luis Castelazo Ayala”, Instituto Mexicano del Seguro Social, México City 01090, Mexico; (L.M.-S.); (G.L.-F.)
| | - Gabriel Lara-Flores
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia N° 4 “Luis Castelazo Ayala”, Instituto Mexicano del Seguro Social, México City 01090, Mexico; (L.M.-S.); (G.L.-F.)
| | - Augusto R. Aguilera-Joaquín
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia No.3, CMN La Raza, Instituto Mexicano del Seguro Social, México City 02990, Mexico; (L.C.-R.); (A.R.A.-J.); (L.S.-G.)
| | - Luisa Sánchez-García
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia No.3, CMN La Raza, Instituto Mexicano del Seguro Social, México City 02990, Mexico; (L.C.-R.); (A.R.A.-J.); (L.S.-G.)
| |
Collapse
|
10
|
Lipid Composition, Digestion, and Absorption Differences among Neonatal Feeding Strategies: Potential Implications for Intestinal Inflammation in Preterm Infants. Nutrients 2021; 13:nu13020550. [PMID: 33567518 PMCID: PMC7914900 DOI: 10.3390/nu13020550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of morbidity and mortality in the neonatal population. Formula feeding is among the many risk factors for developing the condition, a practice often required in the cohort most often afflicted with NEC, preterm infants. While the virtues of many bioactive components of breast milk have been extolled, the ability to digest and assimilate the nutritional components of breast milk is often overlooked. The structure of formula differs from that of breast milk, both in lipid composition and chemical configuration. In addition, formula lacks a critical digestive enzyme produced by the mammary gland, bile salt-stimulated lipase (BSSL). The gastrointestinal system of premature infants is often incapable of secreting sufficient pancreatic enzymes for fat digestion, and pasteurization of donor milk (DM) has been shown to inactivate BSSL, among other important compounds. Incompletely digested lipids may oxidize and accumulate in the distal gut. These lipid fragments are thought to induce intestinal inflammation in the neonate, potentially hastening the development of diseases such as NEC. In this review, differences in breast milk, pasteurized DM, and formula lipids are highlighted, with a focus on the ability of those lipids to be digested and subsequently absorbed by neonates, especially those born prematurely and at risk for NEC.
Collapse
|
11
|
Brink LR, Lönnerdal B. Milk fat globule membrane: the role of its various components in infant health and development. J Nutr Biochem 2020; 85:108465. [PMID: 32758540 DOI: 10.1016/j.jnutbio.2020.108465] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022]
Abstract
Breastfeeding confers many benefits to the breast-fed infant which are reflected by better short-term and long-term outcomes as compared to formula-fed infants. Many components of breast milk are likely to contribute to these favorable outcomes, and there has recently been focus on the milk fat globule membrane (MFGM). This fraction is a heterogenous mixture of proteins (many of them glycosylated), phospholipids, sphingolipids, gangliosides, choline, sialic acid and cholesterol which is lacking in infant formula as milk fat (which is also low in these components) is replaced by vegetable oils. Many of these components have been shown to have biological effects, and there is considerable evidence from preclinical studies and clinical trials that providing bovine MFGM results in improved outcomes, in particular with regard to infections and neurodevelopment. Since bovine MFGM is commercially available, it is possible to add it to infant formula. There are, however, considerable variations in composition among commercial sources of bovine MFGM, and as it is not known which of the individual components provide the various bioactivities, it becomes important to critically review studies to date and to delineate the mechanisms behind the activities observed. In this review, we critically examine the preclinical and clinical studies on MFGM and its components in relation to resistance to infections, cognitive development, establishment of gut microbiota and infant metabolism, and discuss possible mechanisms of action.
Collapse
Affiliation(s)
- Lauren R Brink
- Department of Nutrition, University of California, Davis, 95616
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, 95616.
| |
Collapse
|
12
|
Fontecha J, Brink L, Wu S, Pouliot Y, Visioli F, Jiménez-Flores R. Sources, Production, and Clinical Treatments of Milk Fat Globule Membrane for Infant Nutrition and Well-Being. Nutrients 2020; 12:E1607. [PMID: 32486129 PMCID: PMC7352329 DOI: 10.3390/nu12061607] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022] Open
Abstract
Research on milk fat globule membrane (MFGM) is gaining traction. The interest is two-fold; on the one hand, it is a unique trilayer structure with specific secretory function. On the other hand, it is the basis for ingredients with the presence of phospho- and sphingolipids and glycoproteins, which are being used as food ingredients with valuable functionality, in particular, for use as a supplement in infant nutrition. This last application is at the center of this Review, which aims to contribute to understanding MFGM's function in the proper development of immunity, cognition, and intestinal trophism, in addition to other potential effects such as prevention of diseases including cardiovascular disease, impaired bone turnover and inflammation, skin conditions, and infections as well as age-associated cognitive decline and muscle loss. The phospholipid composition of MFGM from bovine milk is quite like human milk and, although there are some differences due to dairy processing, these do not result in a chemical change. The MFGM ingredients, as used to improve the formulation in different clinical studies, have indeed increased the presence of phospholipids, sphingolipids, glycolipids, and glycoproteins with the resulting benefits of different outcomes (especially immune and cognitive outcomes) with no reported adverse effects. Nevertheless, the precise mechanism(s) of action of MFGM remain to be elucidated and further basic investigation is warranted.
Collapse
Affiliation(s)
- Javier Fontecha
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CIAL, CSIC-UAM), 28049 Madrid, Spain
| | - Lauren Brink
- Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN 47721, USA; (L.B.); (S.W.)
| | - Steven Wu
- Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN 47721, USA; (L.B.); (S.W.)
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods (INAF), Department of Food Sciences, Laval University, Québec, QC G1V 0A6, Canada;
| | - Francesco Visioli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy;
- IMDEA-Food, CEI UAM + CSIC, 28049 Madrid, Spain
| | - Rafael Jiménez-Flores
- Food Science and Technology Department, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
13
|
Ramiro-Cortijo D, Singh P, Liu Y, Medina-Morales E, Yakah W, Freedman SD, Martin CR. Breast Milk Lipids and Fatty Acids in Regulating Neonatal Intestinal Development and Protecting against Intestinal Injury. Nutrients 2020; 12:E534. [PMID: 32092925 PMCID: PMC7071444 DOI: 10.3390/nu12020534] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Human breast milk is the optimal source of nutrition for infant growth and development. Breast milk fats and their downstream derivatives of fatty acids and fatty acid-derived terminal mediators not only provide an energy source but also are important regulators of development, immune function, and metabolism. The composition of the lipids and fatty acids determines the nutritional and physicochemical properties of human milk fat. Essential fatty acids, including long-chain polyunsaturated fatty acids (LCPUFAs) and specialized pro-resolving mediators, are critical for growth, organogenesis, and regulation of inflammation. Combined data including in vitro, in vivo, and human cohort studies support the beneficial effects of human breast milk in intestinal development and in reducing the risk of intestinal injury. Human milk has been shown to reduce the occurrence of necrotizing enterocolitis (NEC), a common gastrointestinal disease in preterm infants. Preterm infants fed human breast milk are less likely to develop NEC compared to preterm infants receiving infant formula. Intestinal development and its physiological functions are highly adaptive to changes in nutritional status influencing the susceptibility towards intestinal injury in response to pathological challenges. In this review, we focus on lipids and fatty acids present in breast milk and their impact on neonatal gut development and the risk of disease.
Collapse
Affiliation(s)
- David Ramiro-Cortijo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Yan Liu
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Esli Medina-Morales
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - William Yakah
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
| | - Steven D. Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Camilia R. Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
14
|
Comitini F, Peila C, Fanos V, Coscia A. The Docosahexanoic Acid: From the Maternal-Fetal Dyad to Early Life Toward Metabolomics. Front Pediatr 2020; 8:538. [PMID: 33102402 PMCID: PMC7555995 DOI: 10.3389/fped.2020.00538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/27/2020] [Indexed: 02/05/2023] Open
Abstract
Docosahexaenoic acid (DHA) is an essential ω-3 long-chain polyunsaturated fatty acid (LCPUFA) and represents the dominant structural fatty acid in the retina and in the brain's gray matter. Due to its active participation in the development of the nervous system, DHA is one of the most studied LCPUFA and is currently considered a critical nutrient during pregnancy and breastfeeding. Increasing evidence in literature suggests that an adequate concentration of DHA is required from the fetal stage through to early life to ensure optimal neurological development. Likewise, many studies in literature demonstrated that an adequate supply of DHA during pregnancy and lactation is essential to promote proper brain development in utero and in early life. Daily supplementation of DHA in newborns has potentially stronger effects compared to maternal supplementation during pregnancy. Supplementation initiated in the second year of life in children born preterm did not result in global cognitive development improvements. Preliminary findings arising from metabolomics has reported that mother's milk and infant formula supplementation of Vitamin D associated with DHA results in a higher antioxidant and protective action, with a possible positive influence on renal function and body fat on preterm infants compared to those receiving only vitamin D. Recent applications of metabolomic studies on newborns may lead to a better understanding of the metabolic process linked to early nutrition and, subsequently, to the development of targeted and personalized nutritional strategies.
Collapse
Affiliation(s)
- Federica Comitini
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University Hospital and University of Cagliari, Monserrato, Italy
| | - Chiara Peila
- Complex Structure Neonatology Unit, Department of Public Health and Paediatric, University of Turin, Turin, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University Hospital and University of Cagliari, Monserrato, Italy
| | - Alessandra Coscia
- Complex Structure Neonatology Unit, Department of Public Health and Paediatric, University of Turin, Turin, Italy
| |
Collapse
|
15
|
Frost BL, Caplan MS. Can Fish Oil Reduce the Incidence of Necrotizing Enterocolitis by Altering the Inflammatory Response? Clin Perinatol 2019; 46:65-75. [PMID: 30771820 DOI: 10.1016/j.clp.2018.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating bowel necrosis that predominantly affects preterm infants and is characterized by an imbalance toward a proinflammatory state. Fish oil or omega-3 long-chain polyunsaturated fatty acids have the potential to modulate inflammation. In this article, the authors examine the evidence in support of fish oil supplementation to alter the inflammatory response and potentially reduce the risk of NEC.
Collapse
Affiliation(s)
- Brandy L Frost
- Department of Pediatrics, NorthShore University HealthSystem, University of Chicago Pritzker School of Medicine, 2650 Ridge Avenue, Walgreen Building Suite 1505, Evanston, IL 60201, USA.
| | - Michael S Caplan
- Department of Pediatrics, NorthShore University HealthSystem, University of Chicago Pritzker School of Medicine, 2650 Ridge Avenue, Walgreen Building Suite 1505, Evanston, IL 60201, USA
| |
Collapse
|
16
|
Polar lipid composition of bioactive dairy co-products buttermilk and butterserum: Emphasis on sphingolipid and ceramide isoforms. Food Chem 2018; 240:67-74. [DOI: 10.1016/j.foodchem.2017.07.091] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 06/20/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022]
|
17
|
Guerin J, Burgain J, Gomand F, Scher J, Gaiani C. Milk fat globule membrane glycoproteins: Valuable ingredients for lactic acid bacteria encapsulation? Crit Rev Food Sci Nutr 2017; 59:639-651. [PMID: 28976212 DOI: 10.1080/10408398.2017.1386158] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The membrane (Milk Fat Globule Membrane - MFGM) surrounding the milk fat globule is becoming increasingly studied for its use in food applications due to proven nutritional and technological properties. This review focuses first on current researches which have been led on the MFGM structure and composition and also on laboratory and industrial purification and isolation methods developed in the last few years. The nutritional, health benefits and techno-functional properties of the MFGM are then discussed. Finally, new techno-functional opportunities of MFGM glycoproteins as a possible ingredient for Lactic Acid Bacteria (LAB) encapsulation are detailed. The ability of MFGM to form liposomes entrapping bioactive compounds has been already demonstrated. One drawback is that liposomes are too small to be used for bacteria encapsulation. For the first time, this review points out the numerous advantages to use MFGM glycoproteins as a protecting, encapsulating matrix for bacteria and especially for LAB.
Collapse
Affiliation(s)
- Justine Guerin
- a LIBio, Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine , 2, av de la Forêt de Haye, BP, Vandœuvre-lès-Nancy , France
| | - Jennifer Burgain
- a LIBio, Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine , 2, av de la Forêt de Haye, BP, Vandœuvre-lès-Nancy , France
| | - Faustine Gomand
- a LIBio, Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine , 2, av de la Forêt de Haye, BP, Vandœuvre-lès-Nancy , France
| | - Joël Scher
- a LIBio, Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine , 2, av de la Forêt de Haye, BP, Vandœuvre-lès-Nancy , France
| | - Claire Gaiani
- a LIBio, Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine , 2, av de la Forêt de Haye, BP, Vandœuvre-lès-Nancy , France
| |
Collapse
|
18
|
Abstract
Fatty acids are critical nutrient regulators of intracellular signaling and influence key pathways including inflammatory responses, hemostasis as well as central nervous system development and function. Preterm birth interrupts the maternal-fetal transfer of essential fatty acids including docosahexaenoic and arachidonic acids, which occurs during the third trimester. Postnatal deficits of these nutrients accrue in preterm infants during the first week and they remain throughout the first months. Due to the regulatory roles of these fatty acids, such deficits contribute an increased risk of developing prematurity-related morbidities including impaired growth and neurodevelopment. The fatty acid contents of parenteral and enteral nutrition are insufficient to meet current recommendations. This chapter summarizes the regulatory roles of fatty acids, current recommendations and limitations of parenteral and enteral nutrition in meeting these recommendations in preterm infants. Suggested areas for research on the roles of fatty acids in preterm infant health are also provided.
Collapse
|
19
|
Schindler T, Sinn JKH, Osborn DA. Polyunsaturated fatty acid supplementation in infancy for the prevention of allergy. Cochrane Database Syst Rev 2016; 10:CD010112. [PMID: 27788565 PMCID: PMC6464137 DOI: 10.1002/14651858.cd010112.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Early dietary intakes may influence the development of allergic disease. It is important to determine if dietary polyunsaturated fatty acids (PUFAs) given as supplements or added to infant formula prevent the development of allergy. OBJECTIVES To determine the effect of higher PUFA intake during infancy to prevent allergic disease. SEARCH METHODS We used the standard search strategy of the Cochrane Neonatal Review group to search the Cochrane Central Register of Controlled Trials (CENTRAL 2015, Issue 9), MEDLINE (1966 to 14 September 2015), EMBASE (1980 to 14 September 2015) and CINAHL (1982 to 14 September 2015). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Randomised and quasi-randomised controlled trials that compared the use of a PUFA with no PUFA in infants for the prevention of allergy. DATA COLLECTION AND ANALYSIS Two review authors independently selected trials, assessed trial quality and extracted data from the included studies. We used fixed-effect analyses. The treatment effects were expressed as risk ratio (RR) with 95% confidence intervals (CI). We used the GRADE approach to assess the quality of evidence. MAIN RESULTS The search found 17 studies that assessed the effect of higher versus lower intake of PUFAs on allergic outcomes in infants. Only nine studies enrolling 2704 infants reported allergy outcomes that could be used in meta-analyses. Of these, there were methodological concerns for eight.In infants up to two years of age, meta-analyses found no difference in incidence of all allergy (1 study, 323 infants; RR 0.96, 95% CI 0.73 to 1.26; risk difference (RD) -0.02, 95% CI -0.12 to 0.09; heterogeneity not applicable), asthma (3 studies, 1162 infants; RR 1.04, 95% CI 0.80 to 1.35, I2 = 0%; RD 0.01, 95% CI -0.04 to 0.05, I2 = 0%), dermatitis/eczema (7 studies, 1906 infants; RR 0.93, 95% CI 0.82 to 1.06, I2 = 0%; RD -0.02, 95% CI -0.06 to 0.02, I2 = 0%) or food allergy (3 studies, 915 infants; RR 0.81, 95% CI 0.56 to 1.19, I2 = 63%; RD -0.02, 95% CI -0.06 to 0.02, I2 = 74%). There was a reduction in allergic rhinitis (2 studies, 594 infants; RR 0.47, 95% CI 0.23 to 0.96, I2 = 6%; RD -0.04, 95% CI -0.08 to -0.00, I2 = 54%; number needed to treat for an additional beneficial outcome (NNTB) 25, 95% CI 13 to ∞).In children aged two to five years, meta-analysis found no difference in incidence of all allergic disease (2 studies, 154 infants; RR 0.69, 95% CI 0.47 to 1.02, I2 = 43%; RD -0.16, 95% CI -0.31 to -0.00, I2 = 63%; NNTB 6, 95% CI 3 to ∞), asthma (1 study, 89 infants; RR 0.45, 95% CI 0.20 to 1.02; RD -0.20, 95% CI -0.37 to -0.02; heterogeneity not applicable; NNTB 5, 95% CI 3 to 50), dermatitis/eczema (2 studies, 154 infants; RR 0.65, 95% CI 0.34 to 1.24, I2 = 0%; RD -0.09 95% CI -0.22 to 0.04, I2 = 24%) or food allergy (1 study, 65 infants; RR 2.27, 95% CI 0.25 to 20.68; RD 0.05, 95% CI -0.07 to 0.16; heterogeneity not applicable).In children aged two to five years, meta-analysis found no difference in prevalence of all allergic disease (2 studies, 633 infants; RR 0.98, 95% CI 0.81 to 1.19, I2 = 36%; RD -0.01, 95% CI -0.08 to 0.07, I2 = 0%), asthma (2 studies, 635 infants; RR 1.12, 95% CI 0.82 to 1.53, I2 = 0%; RD 0.02, 95% CI -0.04 to 0.09, I2 = 0%), dermatitis/eczema (2 studies, 635 infants; RR 0.81, 95% CI 0.59 to 1.09, I2 = 0%; RD -0.04 95% CI -0.11 to 0.02, I2 = 0%), allergic rhinitis (2 studies, 635 infants; RR 1.02, 95% CI 0.83 to 1.25, I2 = 0%; RD 0.01, 95% CI -0.06 to 0.08, I2 = 0%) or food allergy (1 study, 119 infants; RR 0.27, 95% CI 0.06 to 1.19; RD -0.10, 95% CI -0.20 to -0.00; heterogeneity not applicable; NNTB 10, 95% CI 5 to ∞). AUTHORS' CONCLUSIONS There is no evidence that PUFA supplementation in infancy has an effect on infant or childhood allergy, asthma, dermatitis/eczema or food allergy. However, the quality of evidence was very low. There was insufficient evidence to determine an effect on allergic rhinitis.
Collapse
Affiliation(s)
- Tim Schindler
- Royal Hospital for WomenDepartment of Newborn CareBarker StreetRandwickNSWAustralia2031
| | - John KH Sinn
- Royal North Shore Hospital, The University of SydneyDepartment of NeonatologySt. Leonard'sSydneyNew South WalesAustralia2065
| | - David A Osborn
- University of SydneyCentral Clinical School, Discipline of Obstetrics, Gynaecology and NeonatologySydneyNSWAustralia2050
| | | |
Collapse
|
20
|
Fink NH, Collins CT, Gibson RA, Makrides M, Penttila IA. Targeting inflammation in the preterm infant: The role of the omega-3 fatty acid docosahexaenoic acid. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2016. [DOI: 10.1016/j.jnim.2016.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
21
|
Fish oil- and soy oil-based lipid emulsions in neonatal parenteral nutrition: a systematic review and meta-analysis. Eur J Clin Nutr 2016; 70:1106-1115. [PMID: 27142348 DOI: 10.1038/ejcn.2016.69] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 03/12/2016] [Accepted: 03/23/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND/OBJECTIVES The precise role of fish oil-based lipid emulsions (FBL=any lipid emulsion containing any amount of fish oil) and soy oil-based lipid emulsions (SBL=any lipid emulsion not containing fish oil or containing at least 50% soy oil) in neonatal parenteral nutrition (PN) needs to be explored. SUBJECTS/METHODS The PubMed, MedLine and Google Scholar databases were searched for randomised control trials in which PN with FBL and SBL was the only difference between intervention and control groups. Methodological quality was assessed based on the Cochrane handbook for systemic reviews and Jadad's score. Revman 5.2 software was used for meta-analysis. RESULTS Of 420 trials, 25 met the inclusion criteria. The aggregate results showed that both FBLs and SBLs are beneficial. FBLs were associated with significantly lower incidences of cholestasis (relative risk (RR)=0.50; 95% confidence interval (CI)=0.27-0.92, P=0.03) compared with SBLs. FBLs also had a tendency to improve the rates of weight gain, increase in head circumference and the time to regain birth weight (mean difference (MD)=0.24; 95% CI=-0.63-0.15). SBLs were associated with reduced duration of respiratory support (MD=2.22, 95% CI=0.35-4.09, P=0.02) and mortality rate in the first 28 days (RR=1.24, 95% CI=-0.72-2.13). CONCLUSIONS Both FBLs and SBLs are beneficial for neonatal PN. FBLs reduce cholestasis, and SBLs reduce the duration of respiratory support.
Collapse
|
22
|
Hadley KB, Ryan AS, Forsyth S, Gautier S, Salem N. The Essentiality of Arachidonic Acid in Infant Development. Nutrients 2016; 8:216. [PMID: 27077882 PMCID: PMC4848685 DOI: 10.3390/nu8040216] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 01/16/2023] Open
Abstract
Arachidonic acid (ARA, 20:4n-6) is an n-6 polyunsaturated 20-carbon fatty acid formed by the biosynthesis from linoleic acid (LA, 18:2n-6). This review considers the essential role that ARA plays in infant development. ARA is always present in human milk at a relatively fixed level and is accumulated in tissues throughout the body where it serves several important functions. Without the provision of preformed ARA in human milk or infant formula the growing infant cannot maintain ARA levels from synthetic pathways alone that are sufficient to meet metabolic demand. During late infancy and early childhood the amount of dietary ARA provided by solid foods is low. ARA serves as a precursor to leukotrienes, prostaglandins, and thromboxanes, collectively known as eicosanoids which are important for immunity and immune response. There is strong evidence based on animal and human studies that ARA is critical for infant growth, brain development, and health. These studies also demonstrate the importance of balancing the amounts of ARA and DHA as too much DHA may suppress the benefits provided by ARA. Both ARA and DHA have been added to infant formulas and follow-on formulas for more than two decades. The amounts and ratios of ARA and DHA needed in infant formula are discussed based on an in depth review of the available scientific evidence.
Collapse
Affiliation(s)
- Kevin B Hadley
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Alan S Ryan
- Clinical Research Consulting, 9809 Halston Manor, Boynton Beach, FL 33473, USA.
| | - Stewart Forsyth
- School of Medicine, Dentistry & Nursing, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| | - Sheila Gautier
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Norman Salem
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| |
Collapse
|
23
|
Daily Enteral DHA Supplementation Alleviates Deficiency in Premature Infants. Lipids 2016; 51:423-33. [PMID: 26846324 DOI: 10.1007/s11745-016-4130-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
Docosahexaenoic acid (DHA) is an essential fatty acid (FA) important for health and neurodevelopment. Premature infants are at risk of DHA deficiency and circulating levels directly correlate with health outcomes. Most supplementation strategies have focused on increasing DHA content in mother's milk or infant formula. However, extremely premature infants may not reach full feedings for weeks and commercially available parenteral lipid emulsions do not contain preformed DHA, so blood levels decline rapidly after birth. Our objective was to develop a DHA supplementation strategy to overcome these barriers. This double-blind, randomized, controlled trial determined feasibility, tolerability and efficacy of daily enteral DHA supplementation (50 mg/day) in addition to standard nutrition for preterm infants (24-34 weeks gestational age) beginning in the first week of life. Blood FA levels were analyzed at baseline, full feedings and near discharge in DHA (n = 31) or placebo supplemented (n = 29) preterm infants. Term peers (n = 30) were analyzed for comparison. Preterm infants had lower baseline DHA levels (p < 0.0001). Those receiving DHA had a progressive increase in circulating DHA over time (from 3.33 to 4.09 wt% or 2.88 to 3.55 mol%, p < 0.0001) while placebo-supplemented infants (receiving standard neonatal nutrition) had no increase over time (from 3.35 to 3.32 wt% or 2.91 to 2.87 mol%). Although levels increased with additional DHA supplementation, preterm infants still had lower blood DHA levels than term peers (4.97 wt% or 4.31 mol%) at discharge (p = 0.0002). No differences in adverse events were observed between the groups. Overall, daily enteral DHA supplementation is feasible and alleviates deficiency in premature infants.
Collapse
|
24
|
Wijendran V, Brenna JT, Wang DH, Zhu W, Meng D, Ganguli K, Kothapalli KSD, Requena P, Innis S, Walker WA. Long-chain polyunsaturated fatty acids attenuate the IL-1β-induced proinflammatory response in human fetal intestinal epithelial cells. Pediatr Res 2015; 78:626-33. [PMID: 26270575 PMCID: PMC5046822 DOI: 10.1038/pr.2015.154] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/13/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Evidence suggests that excessive inflammation of the immature intestine may predispose premature infants to necrotizing enterocolitis (NEC). We investigated the anti-inflammatory effects of docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (ARA) in human fetal and adult intestinal epithelial cells (IEC) in primary culture. METHODS Human fetal IEC in culture were derived from a healthy fetal small intestine (H4) or resected small intestine of a neonate with NEC (NEC-IEC). Intestinal cell lines Caco2 and NCM460 in culture were used as models for mature IEC. IEC in culture were pretreated with 100 µmol/l palmitic acid (PAL), DHA, EPA, ARA, or ARA+DHA for 48 h and then stimulated with proinflammatory IL-1β. RESULTS DHA significantly attenuated IL-1β induced proinflammatory IL-8 and IL-6 protein and mRNA in fetal H4, NEC-IEC, and mature Caco2, NCM460 IEC, compared to control and PAL treatment. DHA downregulated IL-1R1 (IL-1β receptor) and NFk β1 mRNA expression in fetal and adult IEC. ARA had potent anti-inflammatory effects with lower IL-8 and IL-6 (protein and mRNA) in fetal H4 but not in NEC-IEC or adult IEC. CONCLUSION The present study provides evidence that DHA and ARA may have important anti-inflammatory functions for prevention of NEC in premature infants.
Collapse
Affiliation(s)
- Vasuki Wijendran
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - JT Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Dong Hao Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Weishu Zhu
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Di Meng
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Kriston Ganguli
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | | | - Pilar Requena
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Sheila Innis
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - WA Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| |
Collapse
|
25
|
Munblit D, Boyle RJ, Warner JO. Factors affecting breast milk composition and potential consequences for development of the allergic phenotype. Clin Exp Allergy 2015; 45:583-601. [PMID: 25077553 DOI: 10.1111/cea.12381] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
There is conflicting evidence on the protective role of breastfeeding in relation to allergic sensitization and disease. The factors in breast milk which influence these processes are still unclear and under investigation. We know that colostrum and breast milk contain a variety of molecules which can influence immune responses in the gut-associated lymphoid tissue of a neonate. This review summarizes the evidence that variations in colostrum and breast milk composition can influence allergic outcomes in the infant, and the evidence that maternal and environmental factors can modify milk composition. Taken together, the data presented support the possibility that maternal dietary interventions may be an effective way to promote infant health through modification of breast milk composition.
Collapse
Affiliation(s)
- D Munblit
- Department of Paediatrics, Imperial College London, London, UK; International Inflammation (in-FLAME) Network, of the World Universities Network (WUN)
| | | | | |
Collapse
|
26
|
Nimalaratne C, Wu J. Hen Egg as an Antioxidant Food Commodity: A Review. Nutrients 2015; 7:8274-93. [PMID: 26404361 PMCID: PMC4632414 DOI: 10.3390/nu7105394] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 01/04/2023] Open
Abstract
Intake of antioxidants through diet is known to be important in reducing oxidative damage in cells and improving human health. Although eggs are known for their exceptional, nutritional quality, they are not generally considered as antioxidant foods. This review aims to establish the importance of eggs as an antioxidant food by summarizing the current knowledge on egg-derived antioxidants. Eggs have various natural occurring compounds including the proteins ovalbumin, ovotransferrin and lysozyme in egg white, as well as phosvitin, carotenoids and free aromatic amino acids in egg yolk. Some lipophilic antioxidants such as vitamin E, carotenoids, selenium, iodine and others can be transferred from feed into egg yolk to produce antioxidant-enriched eggs. The bioactivity of egg antioxidants can be affected by food processing, storage and gastrointestinal digestion. Generally thermal processing methods can promote loss of antioxidant properties in eggs due to oxidation and degradation, whereas gastrointestinal digestion enhances the antioxidant properties, due to the formation of new antioxidants (free amino acids and peptides). In summary, in addition to its well-known nutritional contribution to our diet, this review emphasizes the role of eggs as an important antioxidant food.
Collapse
Affiliation(s)
- Chamila Nimalaratne
- Department of Agricultural, Food and Nutritional Science (AFNS), 4-10 Agriculture/Forestry Centre, University of Alberta, Edmonton, AB T6G 2P5, Canada.
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science (AFNS), 4-10 Agriculture/Forestry Centre, University of Alberta, Edmonton, AB T6G 2P5, Canada.
| |
Collapse
|
27
|
Baack ML, Puumala SE, Messier SE, Pritchett DK, Harris WS. What is the relationship between gestational age and docosahexaenoic acid (DHA) and arachidonic acid (ARA) levels? Prostaglandins Leukot Essent Fatty Acids 2015; 100. [PMID: 26205427 PMCID: PMC4554773 DOI: 10.1016/j.plefa.2015.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Long chain polyunsaturated fatty acids (LCPUFA) including docosahexaenoic acid (DHA) and arachidonic acid (ARA) are increasingly transferred from mother to fetus late in pregnancy. Infants born before this transfer is complete are at risk for deficiency. This study determines the relationship between gestational age (GA) and circulating LCPUFA levels to better understand the unique needs of premature infants born at various GAs. Whole blood was collected within the first 7 days of life from 60 preterm (≤34 weeks GA) and 30 term infants (≥38 weeks GA) and FA levels were analyzed. Since concurrent intravenous lipid emulsion can skew composition data, blood LCPUFA concentrations were also measured. Levels were compared among groups, and linear regression models were used to examine the association between FA composition and GA. Preterm infants had significantly lower DHA and ARA levels than term peers, and whether assessed as concentrations or compositions, both directly correlated with GA (p<0.0001). Moreover, FA comparisons suggest that premature infants have impaired synthesis of LCPUFAs from precursors and may require preformed DHA and ARA. This study confirms that essential FA status is strongly related to GA, and that those babies born the earliest are at the greatest risk of LCPUFA deficiency.
Collapse
Affiliation(s)
- Michelle L Baack
- Sanford Children's Hospital, Sioux Falls, SD, USA; Sanford School of Medicine, Sioux Falls, SD, USA; Sanford Health Research Center, Sioux Falls, SD, USA.
| | - Susan E Puumala
- Sanford School of Medicine, Sioux Falls, SD, USA; Sanford Health Research Center, Sioux Falls, SD, USA
| | - Stephen E Messier
- Sanford Children's Hospital, Sioux Falls, SD, USA; Sanford School of Medicine, Sioux Falls, SD, USA
| | | | - William S Harris
- Sanford School of Medicine, Sioux Falls, SD, USA; Sanford Health Research Center, Sioux Falls, SD, USA; OmegaQuant Analytics, LLC, Sioux Falls, SD, USA
| |
Collapse
|
28
|
Modification of Docosahexaenoic Acid Composition of Milk from Nursing Women Who Received Alpha Linolenic Acid from Chia Oil during Gestation and Nursing. Nutrients 2015; 7:6405-24. [PMID: 26247968 PMCID: PMC4555128 DOI: 10.3390/nu7085289] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 05/12/2015] [Accepted: 07/24/2015] [Indexed: 01/29/2023] Open
Abstract
α-Linolenic acid (ALA) is the precursor of docosahexaenoic acid (DHA) in humans, which is fundamental for brain and visual function. Western diet provides low ALA and DHA, which is reflected in low DHA in maternal milk. Chia oil extracted from chia (Salvia hispanica L.), a plant native to some Latin American countries, is high in ALA (up to 60%) and thereby is an alternative to provide ALA with the aim to reduce DHA deficits. We evaluated the modification of the fatty acid profile of milk obtained from Chilean mothers who received chia oil during gestation and nursing. Forty healthy pregnant women (22–35 years old) tabulated for food consumption, were randomly separated into two groups: a control group with normal feeding (n = 21) and a chia group (n = 19), which received 16 mL chia oil daily from the third trimester of pregnancy until the first six months of nursing. The fatty acid profile of erythrocyte phospholipids, measured at six months of pregnancy, at time of delivery and at six months of nursing, and the fatty acid profile of the milk collected during the first six months of nursing were assessed by gas-chromatography. The chia group, compared to the control group, showed (i) a significant increase in ALA ingestion and a significant reduction of linoleic acid (LA) ingestion, no showing modification of arachidonic acid (AA), eicosapentaenoic acid (EPA) and DHA; (ii) a significant increase of erythrocyte ALA and EPA and a reduction of LA. AA and DHA were not modified; (iii) a increased milk content of ALA during the six months of nursing, whereas LA showed a decrease. AA and EPA were not modified, however DHA increased only during the first three months of nursing. Consumption of chia oil during the last trimester of pregnancy and the first three months of nursing transiently increases the milk content of DHA.
Collapse
|
29
|
Beyond building better brains: bridging the docosahexaenoic acid (DHA) gap of prematurity. J Perinatol 2015; 35:1-7. [PMID: 25357095 PMCID: PMC4281288 DOI: 10.1038/jp.2014.195] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/23/2014] [Accepted: 08/26/2014] [Indexed: 12/12/2022]
Abstract
Long-chain polyunsaturated fatty acids (LCPUFA) including docosahexaenoic acid (DHA) are essential for normal vision and neurodevelopment. DHA accretion in utero occurs primarily in the last trimester of pregnancy to support rapid growth and brain development. Premature infants, born before this process is complete, are relatively deficient in this essential fatty acid. Very low birth weight (VLBW) infants remain deficient for a long period of time due to ineffective conversion from precursor fatty acids, lower fat stores and a limited nutritional provision of DHA after birth. In addition to long-term visual and neurodevelopmental risks, VLBW infants have significant morbidity and mortality from diseases specific to premature birth, including bronchopulmonary dysplasia, necrotizing enterocolitis, and retinopathy of prematurity. There is increasing evidence that DHA has protective benefits against these disease states. The aim of this article is to identify the unique needs of premature infants, review the current recommendations for LCPUFA provision in infants and discuss the caveats and innovative new ways to overcome the DHA deficiency through postnatal supplementation, with the long-term goal of improving morbidity and mortality in this at-risk population.
Collapse
|
30
|
Gobbetti T, Ducheix S, le Faouder P, Perez T, Riols F, Boue J, Bertrand-Michel J, Dubourdeau M, Guillou H, Perretti M, Vergnolle N, Cenac N. Protective effects of n-6 fatty acids-enriched diet on intestinal ischaemia/reperfusion injury involve lipoxin A4 and its receptor. Br J Pharmacol 2014; 172:910-23. [PMID: 25296998 DOI: 10.1111/bph.12957] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Long-term intake of dietary fatty acids is known to predispose to chronic inflammation, but their effects on acute intestinal ischaemia/reperfusion (I/R) injury is unknown. The aim of this study was to determine the consequences of a diet rich in n-3 or n-6 polyunsaturated fatty acids (PUFA) on intestinal I/R-induced damage. EXPERIMENTAL APPROACH Mice were fed three different isocaloric diets: a balanced diet used as a control and two different PUFA-enriched diets, providing either high levels of n-3 or of n-6 PUFA. Intestinal injury was evaluated after intestinal I/R. PUFA metabolites were quantitated in intestinal tissues by LC-MS/MS. KEY RESULTS In control diet-fed mice, intestinal I/R caused inflammation and increased COX and lipoxygenase-derived metabolites compared with sham-operated animals. Lipoxin A4 (LxA4 ) was significantly and selectively increased after ischaemia. Animals fed a high n-3 diet did not display a different inflammatory profile following intestinal I/R compared with control diet-fed animals. In contrast, intestinal inflammation was decreased in the I/R group fed with high n-6 diet and level of LxA4 was increased post-ischaemia compared with control diet-fed mice. Blockade of the LxA4 receptor (Fpr2), prevented the anti-inflammatory effects associated with the n-6 rich diet. CONCLUSIONS AND IMPLICATIONS This study indicates that high levels of dietary n-6, but not n-3, PUFAs provides significant protection against intestinal I/R-induced damage and demonstrates that the endogenous production of LxA4 can be influenced by diet.
Collapse
Affiliation(s)
- T Gobbetti
- Inserm, U1043, Toulouse, France; CNRS, U5282, Toulouse, France; Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Université Paul Sabatier, Toulouse, France; WHRI, Queen Mary University, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Zhang P, Lavoie PM, Lacaze-Masmonteil T, Rhainds M, Marc I. Omega-3 long-chain polyunsaturated fatty acids for extremely preterm infants: a systematic review. Pediatrics 2014; 134:120-34. [PMID: 24913791 DOI: 10.1542/peds.2014-0459] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Omega-3 long chain polyunsaturated fatty acid (LCPUFA) exposure can be associated with reduced neonatal morbidities. We systematically review the evidence for the benefits of omega-3 LCPUFAs for reducing neonatal morbidities in extremely preterm infants. METHODS Data sources were PubMed, Embase, Center for Reviews and Dissemination, and the Cochrane Register of Controlled Trials. Original studies were selected that included infants born at <29 weeks' gestation, those published until May 2013, and those that evaluated the relationship between omega-3 LCPUFA supplementation and major adverse neonatal outcomes. Data were extracted on study design and outcome. Effect estimates were pooled. RESULTS Of the 1876 studies identified, 18 randomized controlled trials (RCTs) and 6 observational studies met the defined criteria. No RCT specifically targeted a population of extremely preterm infants. Based on RCTs, omega-3 LCPUFA was not associated with a decreased risk of bronchopulmonary dysplasia in infants overall (pooled risk ratio [RR] 0.97, 95% confidence interval [CI] 0.82-1.13], 12 studies, n = 2809 infants); however, when considering RCTs that include only infants born at ≤32 weeks' gestation, a trend toward a reduction in the risk of bronchopulmonary dysplasia (pooled RR 0.88, 95% CI 0.74-1.05, 7 studies, n = 1156 infants) and a reduction in the risk of necrotizing enterocolitis (pooled RR 0.50, 95% CI 0.23-1.10, 5 studies, n = 900 infants) was observed with LCPUFA. CONCLUSIONS Large-scale interventional studies are required to determine the clinical benefits of omega-3 LCPUFA, specifically in extremely preterm infants, during the neonatal period.
Collapse
Affiliation(s)
- Peiyin Zhang
- Department of Pediatrics, Centre Mère-Enfant, and
| | - Pascal M Lavoie
- Department of Pediatrics, Children's and Women's Health Centre, University of British Columbia, Vancouver, Canada; and
| | | | - Marc Rhainds
- Health Technology Assessment Unit, CHU de Quebec, Laval University, Quebec, Canada
| | | |
Collapse
|
33
|
Vegge A, Thymann T, Lauritzen L, Bering SB, Wiinberg B, Sangild PT. Parenteral lipids and partial enteral nutrition affect hepatic lipid composition but have limited short term effects on formula-induced necrotizing enterocolitis in preterm piglets. Clin Nutr 2014; 34:219-28. [PMID: 24703629 DOI: 10.1016/j.clnu.2014.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 03/07/2014] [Accepted: 03/08/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS Rapid transition from total parenteral nutrition (TPN) to enteral feeding is a risk factor for necrotizing enterocolitis (NEC) in preterm infants. We hypothesized that partial enteral nutrition with colostrum, increased proportion of n-3 polyunsaturated fatty acids (PUFA), or exclusion of lipid in TPN would affect short term NEC sensitivity and liver function. METHODS Preterm piglets were fed for three days after birth: 1) TPN with a standard lipid emulsion (Nutriflex Lipid Plus, TPN control group, n = 19), 2) PN plus bovine colostrum as partial enteral nutrition (PN/COL, n = 18), 3) TPN with fish oil (FO) lipids (Omegaven, TPN/FO, n = 19), or 4) TPN with no lipid (TPN/NL, n = 22). After TPN, piglets were fed formula for two days before tissue collection. RESULTS None of the treatments had consistent effect on NEC incidence (∼40-50% across all groups), intestinal morphology and function, relative to TPN. In the liver, there were no signs of steatosis but PN/COL decreased the n-6 PUFA levels, leading to higher n-3/n-6 ratio, GGT activity, and plasma cholesterol and albumin levels, relative to TPN (all p < 0.05). TPN/FO increased the hepatic n-3 levels and n-3/n-6 ratio. TPN/NL treatment led to decreased hepatic n-6 level, n-3/n-6 ratio and bilirubin, albumin and triglycerides, and lowered blood clotting strength (-30%, TPN/NL vs. TPN/COL, p < 0.05). CONCLUSION Partial enteral nutrition with colostrum, increased n-3 PUFAs in TPN, or removal of lipid from the TPN, all affect hepatic lipids and proteins in preterm neonates. These effects do not translate into improved hepatic function or NEC resistance, at least not short term.
Collapse
Affiliation(s)
- Andreas Vegge
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark; Diabetes Pharmacology, Novo Nordisk A/S, Denmark
| | - Thomas Thymann
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Lotte Lauritzen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Stine B Bering
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Bo Wiinberg
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Haemophilia Pharmacology, Novo Nordisk A/S, Denmark
| | - Per T Sangild
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark.
| |
Collapse
|
34
|
Coursodon-Boyiddle CF, Snarrenberg CL, Adkins-Rieck CK, Bassaganya-Riera J, Hontecillas R, Lawrence P, Brenna JT, Jouni ZE, Dvorak B. Pomegranate seed oil reduces intestinal damage in a rat model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2012; 303:G744-51. [PMID: 22821948 PMCID: PMC3468537 DOI: 10.1152/ajpgi.00248.2012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pomegranate seed oil (PSO), which is the major source of conjugated linolenic acids such as punicic acid (PuA), exhibits strong anti-inflammatory properties. Necrotizing enterocolitis (NEC) is a devastating disease associated with severe and excessive intestinal inflammation. The aim of this study was to evaluate the effects of orally administered PSO on the development of NEC, intestinal epithelial proliferation, and cytokine regulation in a rat model of NEC. Premature rats were divided into three groups: dam fed (DF), formula-fed rats (FF), or rats fed with formula supplemented with 1.5% of PSO (FF + PSO). All groups were exposed to asphyxia/cold stress to induce NEC. Intestinal injury, epithelial cell proliferation, cytokine production, and trefoil factor 3 (Tff3) production were evaluated in the terminal ileum. Oral administration of PSO (FF+PSO) decreased the incidence of NEC from 61 to 26%. Feeding formula with PSO improved enterocyte proliferation in the site of injury. Increased levels of proinflammatory IL-6, IL-8, IL-12, IL-23, and TNF-α in the ileum of FF rats were normalized in PSO-treated animals. Tff3 production in the FF rats was reduced compared with DF but not further affected by the PSO. In conclusion, administration of PSO protects against NEC in the neonatal rat model. This protective effect is associated with an improvement of intestinal epithelial homeostasis and a strong anti-inflammatory effect of PSO on the developing intestinal mucosa.
Collapse
Affiliation(s)
| | - Chelsea L. Snarrenberg
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| | - Camille K. Adkins-Rieck
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| | - Josep Bassaganya-Riera
- 2Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia;
| | - Raquel Hontecillas
- 2Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia;
| | - Peter Lawrence
- 3Division of Nutritional Sciences, Cornell University, Ithaca, New York;
| | - J. Thomas Brenna
- 3Division of Nutritional Sciences, Cornell University, Ithaca, New York;
| | | | - Bohuslav Dvorak
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| |
Collapse
|
35
|
Patel BK, Shah JS. Necrotizing enterocolitis in very low birth weight infants: a systemic review. ISRN GASTROENTEROLOGY 2012; 2012:562594. [PMID: 22997587 PMCID: PMC3444861 DOI: 10.5402/2012/562594] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/19/2012] [Indexed: 01/08/2023]
Abstract
Necrotizing enterocolitis (NEC) is the most common serious gastrointestinal disorder affecting very preterm or very low birth weight infants. The risk is inversely proportional to gestational age and weight at birth. Fetal growth restriction and compromise may be additional specific risk factors. Despite extensive research and animal studies etiopathogenesis, preventive strategies and management options remain controversial. The present paper reviews the literature for recent advances and newer insights for changing epidemiological trends, pathogenesis, role of inflammatory cytokines, and various preventive and management strategies.
Collapse
Affiliation(s)
- Bhoomika K. Patel
- Department of Clinical Pharmacy, Shri Sarvajanik Pharmacy College, Near Arvind Baug, Gujarat, Mehsana 384001, India
| | - Jigna S. Shah
- Department of Clinical Pharmacy, Shri Sarvajanik Pharmacy College, Near Arvind Baug, Gujarat, Mehsana 384001, India
| |
Collapse
|
36
|
Veereman-Wauters G, Staelens S, Rombaut R, Dewettinck K, Deboutte D, Brummer RJ, Boone M, Le Ruyet P. Milk fat globule membrane (INPULSE) enriched formula milk decreases febrile episodes and may improve behavioral regulation in young children. Nutrition 2012; 28:749-52. [DOI: 10.1016/j.nut.2011.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 10/21/2011] [Indexed: 10/14/2022]
|
37
|
Ran-Ressler RR, Khailova L, Arganbright KM, Adkins-Rieck CK, Jouni ZE, Koren O, Ley RE, Brenna JT, Dvorak B. Branched chain fatty acids reduce the incidence of necrotizing enterocolitis and alter gastrointestinal microbial ecology in a neonatal rat model. PLoS One 2011; 6:e29032. [PMID: 22194981 PMCID: PMC3237582 DOI: 10.1371/journal.pone.0029032] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/19/2011] [Indexed: 01/05/2023] Open
Abstract
Introduction Branched chain fatty acids (BCFA) are found in the normal term human newborn's gut, deposited as major components of vernix caseosa ingested during late fetal life. We tested the hypothesis that premature infants' lack of exposure to gastrointestinal (GI) BCFA is associated with their microbiota and risk for necrotizing enterocolitis (NEC) using a neonatal rat model. Methods Pups were collected one day before scheduled birth. The pups were exposed to asphyxia and cold stress to induce NEC. Pups were assigned to one of three experimental treatments. DF (dam-fed) ; Control, hand-fed rat milk substitute ; BCFA, hand-fed rat milk substitute with 20%w/w BCFA. Total fat was equivalent (11%wt) for both the Control and BCFA groups. Cecal microbiota were characterized by 16S rRNA gene pyrosequencing, and intestinal injury, ileal cytokine and mucin gene expression, interleukin-10 (IL-10) peptide immunohistochemistry, and BCFA uptake in ileum phospholipids, serum and liver were assessed. Results NEC incidence was reduced by over 50% in the BCFA group compared to the Control group as assessed in ileal tissue; microbiota differed among all groups. BCFA-fed pups harbored greater levels of BCFA-associated Bacillus subtilis and Pseudomonas aeruginosa compared to Controls. Bacillus subtilis levels were five-fold greater in healthy pups compared to pups with NEC. BCFA were selectively incorporated into ileal phospholipids, serum and liver tissue. IL-10 expression increased three-fold in the BCFA group versus Controls and no other inflammatory or mucosal mRNA markers changed. Conclusion At constant dietary fat level, BCFA reduce NEC incidence and alter microbiota composition. BCFA are also incorporated into pup ileum where they are associated with enhanced IL-10 and may exert other specific effects.
Collapse
Affiliation(s)
- Rinat R. Ran-Ressler
- Division of Nutritional Sciences, Savage Hall, Cornell University, Ithaca, New York, United States of America
| | - Ludmila Khailova
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | - Kelly M. Arganbright
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | | | - Zeina E. Jouni
- Mead Johnson Nutrition, Evansville, Indiana, United States of America
| | - Omry Koren
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - Ruth E. Ley
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - J. Thomas Brenna
- Division of Nutritional Sciences, Savage Hall, Cornell University, Ithaca, New York, United States of America
- * E-mail: (JTB); (BD)
| | - Bohuslav Dvorak
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
- * E-mail: (JTB); (BD)
| |
Collapse
|
38
|
Janvier A, Beaumier L, Barrington KJ. Intestinal absorption of lipid emulsion in premature infants: a pilot study. Neonatology 2011; 100:248-52. [PMID: 21701214 DOI: 10.1159/000326079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 02/10/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND Adequate nutritional intake is essential in the very-low-birth-weight infant, but difficult to achieve in the first few postnatal days. Can lipids be given enterally in the first few days of life in sick preterm infants? OBJECTIVE To determine tolerance and absorption of lipid emulsion when fed enterally to very-low-birth-weight infants. DESIGN/METHODS Infants had a birth weight <1,500 g, an appropriate weight for gestational age, and were receiving parenteral nutrition. We performed a progressive series of studies, enrolling 5 infants in each group. Group 1 infants were fed enteral lipid emulsion at 1 g/kg/day for 4 days, starting when 60 ml/kg/day of breast milk was tolerated enterally. Simultaneously, a matched control group which received no oral lipid emulsion was enrolled. We then enrolled group 2 infants who were fed 3 g/kg/day with the same protocol as group 1. Group 3 infants were fed enteral lipid emulsion starting in the first 72 h of life. The infants were fed 1, 2 and 3 g/kg/day subsequently for 48 h each. Fat absorption was measured. RESULTS Gestational age was 24.6-30.8 weeks and birth weight was 620-1,400 g. One infant (group 1) developed necrotizing enterocolitis 1 week after the study. There were no other adverse clinical findings. On average, enteral lipid emulsion was started on day 8 of life in groups 1 and 2, and on day 2 in group 3. The intestinal lipid absorption was 93.6% (min. = 76%). There was no difference in fat absorption between the 4 groups (p > 0.05). CONCLUSIONS Lipid emulsions are an isotonic high-calorie source which can be given safely enterally instead of intravenously in the immediate neonatal period of very-low-birth-weight infants without clinical adverse effects and with almost complete absorption. There are potential advantages to oral administration of a lipid emulsion starting in early life which require further investigation.
Collapse
Affiliation(s)
- A Janvier
- Department of Pediatrics, CHU Sainte Justine, University of Montreal, Montreal, Quebec, Canada
| | | | | |
Collapse
|
39
|
Szajewska H, Makrides M. Is Early Nutrition Related to Short-Term Health and Long-Term Outcome? ANNALS OF NUTRITION AND METABOLISM 2011; 58 Suppl 1:38-48. [DOI: 10.1159/000323465] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
40
|
Siggers RH, Siggers J, Thymann T, Boye M, Sangild PT. Nutritional modulation of the gut microbiota and immune system in preterm neonates susceptible to necrotizing enterocolitis. J Nutr Biochem 2010; 22:511-21. [PMID: 21193301 DOI: 10.1016/j.jnutbio.2010.08.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 08/23/2010] [Indexed: 02/07/2023]
Abstract
The gastrointestinal inflammatory disorder, necrotizing enterocolitis (NEC), is among the most serious diseases for preterm neonates. Nutritional, microbiological and immunological dysfunctions all play a role in disease progression but the relationship among these determinants is not understood. The preterm gut is very sensitive to enteral feeding which may either promote gut adaptation and health, or induce gut dysfunction, bacterial overgrowth and inflammation. Uncontrolled inflammatory reactions may be initiated by maldigestion and impaired mucosal protection, leading to bacterial overgrowth and excessive nutrient fermentation. Tumor necrosis factor alpha, toll-like receptors and heat-shock proteins are identified among the immunological components of the early mucosal dysfunction. It remains difficult, however, to distinguish the early initiators of NEC from the later consequences of the disease pathology. To elucidate the mechanisms and identify clinical interventions, animal models showing spontaneous NEC development after preterm birth coupled with different forms of feeding may help. In this review, we summarize the literature and some recent results from studies on preterm pigs on the nutritional, microbial and immunological interactions during the early feeding-induced mucosal dysfunction and later NEC development. We show that introduction of suboptimal enteral formula diets, coupled with parenteral nutrition, predispose to disease, while advancing amounts of mother's milk from birth (particularly colostrum) protects against disease. Hence, the transition from parenteral to enteral nutrition shortly after birth plays a pivotal role to secure gut growth, digestive maturation and an appropriate response to bacterial colonization in the sensitive gut of preterm neonates.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/microbiology
- Gastrointestinal Tract/growth & development
- Gastrointestinal Tract/immunology
- Gastrointestinal Tract/microbiology
- Heat-Shock Proteins/metabolism
- Humans
- Immune System/immunology
- Infant Nutritional Physiological Phenomena
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/immunology
- Infant, Premature, Diseases/microbiology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/microbiology
- Intestine, Small/metabolism
- Metagenome/physiology
Collapse
Affiliation(s)
- Richard H Siggers
- Department of Human Nutrition, Faculty of Life Sciences, University of Copenhagen, 30 Rolighedsvej, DK-1958 Frederiksberg C, Denmark
| | | | | | | | | |
Collapse
|
41
|
Ehehalt R, Braun A, Karner M, Füllekrug J, Stremmel W. Phosphatidylcholine as a constituent in the colonic mucosal barrier--physiological and clinical relevance. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:983-93. [PMID: 20595010 DOI: 10.1016/j.bbalip.2010.05.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Revised: 05/21/2010] [Accepted: 05/24/2010] [Indexed: 02/09/2023]
Abstract
Phosphatidylcholine (PC) is an important constituent of the gastrointestinal tract. PC molecules are not only important in intestinal cell membranes but also receiving increasing attention as protective agents in the gastrointestinal barrier. They are largely responsible for establishing the hydrophobic surface of the colon. Decreased phospholipids in colonic mucus could be linked to the pathogenesis of ulcerative colitis, a chronic inflammatory bowel disease. Clinical studies revealed that therapeutic addition of PC to the colonic mucus of these patients alleviated the inflammatory activity. This positive role is still elusive, however, we hypothesized that luminal PC has two possible functions: first, it is essential for surface hydrophobicity, and second, it is integrated into the plasma membrane of enterocytes and it modulates the signaling state of the mucosa. The membrane structure and lipid composition of cells is a regulatory component of the inflammatory signaling pathways. In this perspective, we will shortly summarize what is known about the localization and protective properties of PC in the colonic mucosa before turning to its evident medical importance. We will discuss how PC contributes to our understanding of the pathogenesis of ulcerative colitis and how reinforcing the luminal phospholipid monolayer can be used as a therapeutic concept in humans.
Collapse
Affiliation(s)
- Robert Ehehalt
- Department of Gastroenterology, University hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
42
|
Enteral glutamine and/or arginine supplementation have favorable effects on oxidative stress parameters in neonatal rat intestine. J Pediatr Gastroenterol Nutr 2009; 49:85-9. [PMID: 19503000 DOI: 10.1097/mpg.0b013e318198cd36] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE To investigate and compare the effects of enteral glutamine and arginine supply on lipid peroxidation and antioxidant enzyme levels in the small intestine of healthy breast-fed rats. MATERIALS AND METHODS The study comprised 40 newborn Sprague-Dawley rats born to 5 mother rats. Newborn rats were randomly divided into 4 groups. Starting from day 1 until day 21, group I received only breast milk; group II received breast milk and 200 mg/kg/day oral glutamine; group III received breast milk and 200 mg/kg/day oral arginine; and group IV received breast milk, 200 mg/kg/day glutamine, and 200 mg/kg/day arginine. Malondialdehyde levels and glutathione peroxidase (GPx) and superoxide dismutase activities were measured. RESULTS The lowest malondialdehyde levels were found in group II (P = 0.0001). Superoxide dismutase activity was found to be significantly higher in group II than group I (P < 0.001). Of the 4 groups, GPx activity was highest in group IV. GPx activity in group II was significantly higher than in group I (P = 0.001) or group III (P = 0.001). GPx activity was higher in group IV than in group I (P = 0.001) or group III (P = 0.001). CONCLUSIONS Enteral glutamine alone or in the presence of arginine has favorable effects on oxidative stress not only in experimental models of hypoxia-reoxygenation, but also in healthy newborn rats. This suggests that in premature neonates with insufficient oxidative resistance, glutamine and arginine supplementation may help prevent necrotizing enterocolitis.
Collapse
|
43
|
Petrosyan M, Guner YS, Williams M, Grishin A, Ford HR. Current concepts regarding the pathogenesis of necrotizing enterocolitis. Pediatr Surg Int 2009; 25:309-18. [PMID: 19301015 DOI: 10.1007/s00383-009-2344-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2009] [Indexed: 02/07/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease that predominantly affects premature neonates. The mortality associated with NEC has not changed appreciably over the past several decades. The underlying etiology of NEC remains elusive, although bacterial colonization of the gut, formula feeding, and perinatal stress have been implicated as putative risk factors. The disease is characterized by massive epithelial destruction, which results in gut barrier failure. The exact molecular and cellular mechanisms involved in this complex disease are poorly understood. Recent studies have provided significant insight into our understanding of the pathogenesis of NEC. Endogenous mediators such as prostanoids, cyclooxygenases, and nitric oxide may play a role in the development of gut barrier failure. Understanding the structural architecture of the gut barrier and the cellular mechanisms that are responsible for gut epithelial damage could lead to the development of novel diagnostic, prophylactic and therapeutic strategies in NEC.
Collapse
Affiliation(s)
- Mikael Petrosyan
- Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, 4650 Sunset Blvd, Mailstop #72, Los Angeles, CA 90027, USA
| | | | | | | | | |
Collapse
|
44
|
Drenckpohl D, McConnell C, Gaffney S, Niehaus M, Macwan KS. Randomized trial of very low birth weight infants receiving higher rates of infusion of intravenous fat emulsions during the first week of life. Pediatrics 2008; 122:743-51. [PMID: 18829797 DOI: 10.1542/peds.2007-2282] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE The goal was to determine whether very low birth weight infants could tolerate higher rates of infusion of intravenous fat emulsion during the first week of life and maintain their serum triglyceride levels at <or=200 mg/dL. METHODS This was a randomized, controlled trial of 110 infants who were classified as appropriate for gestational age and had birth weights between 750 g and 1500 g. The primary clinical outcome was serum triglyceride levels; secondary outcomes also were monitored. RESULTS One hundred infants completed the study (experimental group: N = 48; control group: N = 52). Infants in the experimental group had significantly higher energy intake for the entire 7-day study period and achieved 90 kcal/kg per day (1 kcal = 4.184 kJ) significantly sooner (7.38 +/- 3.381 days vs 9.44 +/- 3.578 days). Triglyceride levels for infants in the experimental group remained significantly higher for the first 5 days of life. Fifteen percent of infants in the experimental group but only 4% of infants in the control group developed hypertriglyceridemia. Ten percent of infants in the control group but no infants in the experimental group required insulin therapy. Forty-two percent of infants in the experimental group and 17% of infants in the control group remained at >or=10th percentile for weight for age. Fourteen percent of infants in the control group but no infants in the experimental group developed necrotizing enterocolitis. Twenty-three percent of infants in the control group but only 6% of infants in the experimental group developed retinopathy of prematurity. There were no significant differences in other outcomes. CONCLUSIONS Very low birth weight infants can tolerate higher rates of infusion of intravenous fat emulsion solutions during the first week of life without significant adverse events.
Collapse
Affiliation(s)
- Douglas Drenckpohl
- Neonatal Intensive Care Unit, Children's Hospital of Illinois, Third Order of St Francis Medical Center, Peoria, IL 61637, USA.
| | | | | | | | | |
Collapse
|
45
|
Lu J, Caplan MS, Li D, Jilling T. Polyunsaturated fatty acids block platelet-activating factor-induced phosphatidylinositol 3 kinase/Akt-mediated apoptosis in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2008; 294:G1181-90. [PMID: 18356536 PMCID: PMC2692041 DOI: 10.1152/ajpgi.00343.2007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have shown earlier that platelet-activating factor (PAF) causes apoptosis in enterocytes via a mechanism that involves Bax translocation to mitochondria, followed by caspase activation and DNA fragmentation. Herein we report that, in rat small intestinal epithelial cells (IEC-6), these downstream apoptotic effects are mediated by a PAF-induced inhibition of the phosphatidylinositol 3-kinase (PI 3-kinase)/protein kinase B (Akt) signaling pathway. Treatment with PAF results in rapid dephosphorylation of Akt, phosphoinositide-dependent kinase-1, and the YXXM p85 binding motif of several proteins and redistribution of Akt-pleckstrin homology domain-green fluorescent protein, i.e., an in vivo phosphatidylinositol (3,4,5)-trisphosphate sensor, from membrane to cytosol. The proapoptotic effects of PAF were inhibited by both n-3 and n-6 polyunsaturated fatty acids but not by a saturated fatty acid palmitate. Indomethacin, an inhibitor of prostaglandin biosynthesis, did not influence the baseline or PAF-induced apoptosis, but 2-bromopalmitate, an inhibitor of protein palmitoylation, inhibited all of the proapoptotic effects of PAF. Our data strongly suggest that an inhibition of the PI 3-kinase/Akt signaling pathway is the main mechanism of PAF-induced apoptosis in enterocytes and that polyunsaturated fatty acids block this mechanism very early in the signaling cascade independently of any effect on prostaglandin synthesis, and probably directly via an effect on protein palmitoylation.
Collapse
Affiliation(s)
- Jing Lu
- Evanston Northwestern Healthcare Research Institute, Evanston,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinios
| | - Michael S. Caplan
- Evanston Northwestern Healthcare Research Institute, Evanston,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinios
| | - Dan Li
- Evanston Northwestern Healthcare Research Institute, Evanston
| | - Tamas Jilling
- Evanston Northwestern Healthcare Research Institute, Evanston,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinios
| |
Collapse
|
46
|
Dewettinck K, Rombaut R, Thienpont N, Le TT, Messens K, Van Camp J. Nutritional and technological aspects of milk fat globule membrane material. Int Dairy J 2008. [DOI: 10.1016/j.idairyj.2007.10.014] [Citation(s) in RCA: 380] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
47
|
Schnabl KL, Aerde JEV, Thomson ABR, Clandinin MT. Necrotizing enterocolitis: A multifactorial disease with no cure. World J Gastroenterol 2008; 14:2142-61. [PMID: 18407587 PMCID: PMC2703838 DOI: 10.3748/wjg.14.2142] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Necrotizing enterocolitis is an inflammatory bowel disease of neonates with significant morbidity and mortality in preterm infants. Due to the multifactorial nature of the disease and limitations in disease models, early diagnosis remains challenging and the pathogenesis elusive. Although preterm birth, hypoxic-ischemic events, formula feeding, and abnormal bacteria colonization are established risk factors, the role of genetics and vasoactive/inflammatory mediators is unclear. Consequently, treatments do not target the specific underlying disease processes and are symptomatic and surgically invasive. Breast-feeding is the most effective preventative measure. Recent advances in the prevention of necrotizing enterocolitis have focused on bioactive nutrients and trophic factors in human milk. Development of new disease models including the aspect of prematurity that consistently predisposes neonates to the disease with multiple risk factors will improve our understanding of the pathogenesis and lead to discovery of innovative therapeutics.
Collapse
|
48
|
Smithers LG, Gibson RA, McPhee A, Makrides M. Effect of long-chain polyunsaturated fatty acid supplementation of preterm infants on disease risk and neurodevelopment: a systematic review of randomized controlled trials. Am J Clin Nutr 2008; 87:912-20. [PMID: 18400714 DOI: 10.1093/ajcn/87.4.912] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Supplementation of preterm formulas with long-chain polyunsaturated fatty acids (LCPUFAs) is based on their effectiveness to increase blood status and improve visual outcomes. Dispute remains over their efficacy on global development. OBJECTIVE The objective was to compare the effects of LCPUFA-supplemented with those of control formulas on neurodevelopment and diseases associated with prematurity. DESIGN We systematically reviewed randomized controlled trials involving preterm infants that tested LCPUFA-supplemented formulas. The weighted mean differences (WMDs) in neurodevelopmental scores and relative risk (RR) of disease were calculated to compare infants fed LCPUFA-supplemented formula with those fed control formula. RESULTS No clear differences in Bayley Scales of Infant Development (BSID) scores were observed between groups. Mental development of LCPUFA-supplemented infants was 3.4 points higher than that of control infants with BSID version II (WMD: 3.44; 95% CI: 0.56, 6.31; P = 0.02; n = 879), although it was driven by 2 trials with large effect sizes and wide CIs. Psychomotor development was lower in supplemented infants tested with BSID version I (WMD: -7.99; 95% CI: -14.00, -1.99; P = 0.009; n = 87); however, it was limited by small sample sizes. No differences in the RR of sepsis (1.08; 95% CI: 0.79, 1.46; P = 0.63; n = 1333) or in the RR of necrotizing enterocolitis (1.13; 95% CI: 0.62, 2.04; P = 0.69; n = 1333) were found. Similarly, the risks of retinopathy of prematurity, intraventricular hemorrhage, and bronchopulmonary dysplasia were not different between groups, but data were limited by small sample sizes and trials with an increased risk of bias. CONCLUSIONS LCPUFA-supplemented formula does not alter the risk of NEC or sepsis. Further work is needed to determine the extent of benefit of LCPUFA-supplemented formula on the mental development of preterm infants.
Collapse
Affiliation(s)
- Lisa G Smithers
- Child Health Research Institute, Child, Youth and Women's Health Service, North Adelaide, Australia
| | | | | | | |
Collapse
|
49
|
Frost BL, Jilling T, Caplan MS. The importance of pro-inflammatory signaling in neonatal necrotizing enterocolitis. Semin Perinatol 2008; 32:100-6. [PMID: 18346533 PMCID: PMC2362144 DOI: 10.1053/j.semperi.2008.01.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Despite modern medical advances, necrotizing enterocolitis (NEC) remains a significant cause of morbidity and mortality in neonatal intensive care units, affecting 10% of premature neonates born weighing less than 1500 g. Although many advances have been made in the understanding of NEC, the etiology and pathophysiology remain incompletely understood, and treatment is limited to supportive care. In recent years, many studies have evaluated the inflammatory cascade that is central to the disease process, and research is ongoing into strategies to prevent and/or ameliorate neonatal NEC. In this review, we examine the key points in the signaling pathways involved in NEC, and potential strategies for prevention and treatment of this dreaded disease.
Collapse
|
50
|
Affiliation(s)
- Philip C Calder
- Institute of Human Nutrition, School of Medicine, University of Southampton, Bassett Crescent East Southampton SO16 7PX UK.
| |
Collapse
|